101
|
Wang YM, Wang WC, Pan Y, Zeng L, Wu J, Wang ZB, Zhuang XL, Li ML, Cooper DN, Wang S, Shao Y, Wang LM, Fan YY, He Y, Hu XT, Wu DD. Regional and aging-specific cellular architecture of non-human primate brains. Genome Med 2025; 17:41. [PMID: 40296047 PMCID: PMC12038948 DOI: 10.1186/s13073-025-01469-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Deciphering the functionality and dynamics of brain networks across different regions and age groups in non-human primates (NHPs) is crucial for understanding the evolution of human cognition as well as the processes underlying brain pathogenesis. However, systemic delineation of the cellular composition and molecular connections among multiple brain regions and their alterations induced by aging in NHPs remain largely unresolved. METHODS In this study, we performed single-nucleus RNA sequencing on 39 samples collected from 10 brain regions of two young and two aged rhesus macaques using the DNBelab C4 system. Validation of protein expression of signatures specific to particular cell types, brain regions, and aging was conducted through a series of immunofluorescence and immunohistochemistry staining experiments. Loss-of-function experiments mediated by short hairpin RNA (shRNA) targeting two age-related genes (i.e., VSNL1 and HPCAL4) were performed in U251 glioma cells to verify their aging effects. Senescence-associated beta-galactosidase (SA-β-gal) staining and quantitative PCR (qPCR) of senescence marker genes were employed to assess cellular senescence in U251 cells. RESULTS We have established a large-scale cell atlas encompassing over 330,000 cells for the rhesus macaque brain. Our analysis identified numerous gene expression signatures that were specific to particular cell types, subtypes, brain regions, and aging. These datasets greatly expand our knowledge of primate brain organization and highlight the potential involvement of specific molecular and cellular components in both the regionalization and functional integrity of the brain. Our analysis also disclosed extensive transcriptional alterations and cell-cell connections across brain regions in the aging macaques. Finally, by examining the heritability enrichment of human complex traits and diseases, we determined that neurological traits were significantly enriched in neuronal cells and multiple regions with aging-relevant gene expression signatures, while immune-related traits exhibited pronounced enrichment in microglia. CONCLUSIONS Taken together, our study presents a valuable resource for investigating the cellular and molecular architecture of the primate nervous system, thereby expanding our understanding of the mechanisms underlying brain function, aging, and disease.
Collapse
Affiliation(s)
- Yun-Mei Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wen-Chao Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Yongzhang Pan
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lin Zeng
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jing Wu
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Zheng-Bo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650107, China
| | - Xiao-Lin Zhuang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Ming-Li Li
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - David N Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sheng Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Yunnan Key Laboratory of Biodiversity Information, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yong Shao
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Li-Min Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Ying-Yin Fan
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Yonghan He
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xin-Tian Hu
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
| | - Dong-Dong Wu
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
- Kunming Natural History Museum of Zoology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.
| |
Collapse
|
102
|
Li C, Liu W, Fu A, Yang H, Yi G. Potential therapeutic strategies targeting efferocytosis for inflammation resolution and tissue repair in inflammatory bowel disease. Cell Immunol 2025; 411-412:104957. [PMID: 40315792 DOI: 10.1016/j.cellimm.2025.104957] [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: 08/21/2024] [Revised: 04/11/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025]
Abstract
Efferocytosis, the process by which apoptotic cells (ACs) are recognized and cleared by phagocytes, is a critical mechanism in maintaining intestinal immune homeostasis and promoting the resolution of inflammation. Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), is characterized by chronic intestinal inflammation, wherein defective efferocytosis contributes to the accumulation of ACs, secondary necrosis, and sustained mucosal damage. This review delineates the molecular mechanisms underlying efferocytosis and systematically examines its functional roles across five key intestinal phagocytic cell types: macrophages, dendritic cells (DCs), neutrophils, intestinal epithelial cells (IECs), and Paneth cells (PCs). Particular emphasis is placed on the dysregulation of efferocytosis capacity in IBD pathogenesis and the consequences of impaired apoptotic cell clearance in both professional and non-professional phagocytes. Furthermore, we evaluate emerging therapeutic strategies designed to restore or enhance efferocytosis, including modulation of macrophage polarization, LC3-associated phagocytosis pathways, nanotechnology-enabled delivery systems, and stem cell-based interventions. A comprehensive understanding of cell-type-specific efferocytosis in the intestinal microenvironment offers promising directions for the development of targeted, inflammation-resolving therapies for IBD.
Collapse
Affiliation(s)
- Chaoquan Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Wanting Liu
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Aoni Fu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Haotian Yang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Guanghui Yi
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
103
|
Pan YZ, Chen WT, Jin HR, Liu Z, Gu YY, Wang XR, Wang J, Lin JJ, Zhou Y, Xu LM. Correlation between the interleukin-36 subfamily and gut microbiota in patients with liver cirrhosis: Implications for gut-liver axis imbalance. World J Hepatol 2025; 17:105660. [PMID: 40308824 PMCID: PMC12038412 DOI: 10.4254/wjh.v17.i4.105660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Liver cirrhosis (LC) affect millions of people worldwide. The pathogenesis of cirrhosis involves complex interactions between immune responses and gut microbiota. Recent studies have highlighted the role of the interleukin-36 (IL-36) subfamily in inflammation and immune regulation. However, the relationship between serum IL-36 subfamily levels and gut microbiota in cirrhosis patients remains unclear. This study aimed to explore the clinical significance of serum IL-36 subfamily levels and their association with gut microbiota in cirrhosis patients. AIM To explore the clinical significance of serum IL-36 subfamily levels and their relationship with gut microbiota among cirrhosis patients. METHODS Sixty-one cirrhosis patients were enrolled from Lihuili Hospital of Ningbo University from May 2022 to November 2023 as the LC group and 29 healthy volunteers as the healthy control (HC) group. The serum expressions of IL-36α, IL-36β, IL-36γ, IL-36Ra, and IL-38 were measured through ELISA, while 16S rRNA gene sequencing was employed to rate microbial community in human fecal samples. RESULTS The serum levels of IL-36α, IL-36γ, IL-36Ra, and IL-38 in the LC group remarkably exceeded those in the HC group (P < 0.05). IL-36α, IL-36γ, and IL-38 were related positively to the Child-Pugh score (P < 0.05) and prominently exceeded those in the Child-Pugh C group (P < 0.05). The absolute abundance of harmful bacteria (Bacteroides, Bifidobacterium, Faecalibacterium) remarkably rose, while the beneficial bacteria (Firmicutes, Bacteroides, Escherichia-Shigella) notably decreased in the LC group (P < 0.05). IL-36α, IL-36γ, and IL-38 related positively to Lactobacillus (P < 0.05), while IL-38 negatively related to Fusicatenibacter (P < 0.05). CONCLUSION IL-36γ and IL-38 show promise as potential biomarkers for LC progression, but further validation is required.
Collapse
Affiliation(s)
- Yi-Zhi Pan
- Department of Infectious Diseases and Liver Diseases, Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
- Department of Infectious Diseases and Liver Diseases, People's Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Wan-Ting Chen
- Department of Infectious Diseases and Liver Diseases, Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
- Department of Rheumatology and Immunology, Ningbo Hangzhou Bar Hospital, Ningbo 315000, Zhejiang Province, China
| | - Hao-Ran Jin
- Department of Infectious Diseases and Liver Diseases, Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Zhen Liu
- Department of Infectious Diseases and Liver Diseases, Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Ying-Ying Gu
- Department of Infectious Diseases and Liver Diseases, Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Xin-Ruo Wang
- Department of Infectious Diseases and Liver Diseases, People's Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Jue Wang
- Department of Infectious Diseases and Liver Diseases, Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Jing-Jing Lin
- Department of Infectious Diseases and Liver Diseases, Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Yan Zhou
- Department of Infectious Diseases and Liver Diseases, Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
| | - Lan-Man Xu
- Department of Infectious Diseases and Liver Diseases, Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
- Department of Infectious Diseases and Liver Diseases, People's Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China.
| |
Collapse
|
104
|
Zhang Z, Wang X, Zhao C, Zhu H, Liao X, Tsai HI. STING and metabolism-related diseases: Roles, mechanisms, and applications. Cell Signal 2025; 132:111833. [PMID: 40294833 DOI: 10.1016/j.cellsig.2025.111833] [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: 01/23/2025] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
The stimulator of interferon genes (STING) pathway plays a critical role in innate immunity, acting as a central mediator that links cytosolic DNA sensing to inflammatory signaling. STING not only responds to cellular metabolic states but also actively regulates key metabolic processes, including glycolysis, lipid metabolism, and redox balance. This bidirectional interaction underscores the existence of a dynamic feedback mechanism between STING signaling and metabolic pathways, which is essential for maintaining cellular homeostasis. This review provides a comprehensive analysis, beginning with an in-depth overview of the classical STING signaling pathway, followed by a detailed examination of its reciprocal regulation of various metabolic pathways. Additionally, it explores the role and mechanisms of STING signaling in metabolic disorders, including obesity, diabetes, and atherosclerosis. By integrating these insights into the mutual regulation between STING and its metabolism, novel therapeutic strategies targeting this pathway in metabolic diseases have been proposed.
Collapse
Affiliation(s)
- Zhengyang Zhang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xirui Wang
- Department of Biomedical Engineering, School of Medical Imaging, Xuzhou Medical University, Xuzhou 221000, China
| | - Chuangchuang Zhao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Xiang Liao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China.
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
105
|
Yang T, Luo L, Luo X, Liu X. Metabolic crosstalk and therapeutic interplay between diabetes and hyperuricemia. Diabetes Res Clin Pract 2025; 224:112204. [PMID: 40294652 DOI: 10.1016/j.diabres.2025.112204] [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: 03/10/2025] [Revised: 04/20/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
Hyperuricemia and diabetes mellitus (DM) are prevalent metabolic disorders with high comorbidity, imposing a substantial global public health burden. Their coexistence is not merely additive but synergistic, exacerbating metabolic dysregulation through mechanisms such as insulin resistance and β-cell apoptosis, ultimately establishing a vicious cycle. Both disorders induce acute and chronic damage to vital organs, particularly the cardiovascular, renal systems. Hyperuricemia aggravates diabetic complications, notably diabetic cardiomyopathy, nephropathy and retinopathy via oxidative stress, inflammation, and metabolic dysregulation.Current urate-lowering therapies (ULTs), such as xanthine oxidase inhibitors and urate transporter 1 (URAT1, also known as SLC22A12) antagonists, demonstrate potential benefits in ameliorating diabetic complications but face challenges including safety concerns and dose adjustments. Similarly, several glucose-lowering drugs also exhibit the benefits of improving hyperuricemia. This review summarizes the metabolic crosstalk and therapeutic interplay between hyperuricemia and DM, examines the pathogenic role of uric acid in diabetic complications, and discusses the benefits and challenges of existing ULTs and glucose-lowering drugs in disrupting this cycle of metabolic dysregulation and concurrent organ damage. We hope our findings deepen the comprehension of the intricate metabolic crosstalk between glucose and urate homeostasis, providing novel therapeutic insights for patients with comorbid DM and hyperuricemia.
Collapse
Affiliation(s)
- Tianshu Yang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 Hubei, China
| | - Lingyun Luo
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030 Hubei, China
| | - Xuelian Luo
- Department of Oncology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Xiaolei Liu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030 Hubei, China.
| |
Collapse
|
106
|
Di Meo C, Tisi A, Lizzi AR, Palaniappan S, Pulcini F, Cinque B, Delle Monache S, Nazarè M, Hsu E, Rapino C, Maccarrone M. Development of a human RPE In vitro model with AMD-like features reveals blue light-induced modulation of the endocannabinoid system. Biochem Biophys Res Commun 2025; 767:151896. [PMID: 40318376 DOI: 10.1016/j.bbrc.2025.151896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Blue light (BL) is a known risk factor for age-related macular degeneration (AMD), a retinal pathology where damage to the retinal pigment epithelium (RPE) is one of the earliest events. While the endocannabinoid system (ECS) is implicated in various physio-pathological conditions of the retina, its role in BL-injured RPE has not yet been addressed. To fill this gap, we developed an in vitro model of BL-induced human RPE damage showing key features of AMD: cytotoxicity, cell cycle arrest, oxidative stress, inflammation, and cellular senescence. Notably, our model demonstrates modulation of gene and protein expression of specific ECS elements, particularly cannabinoid receptors 1 and 2 (CB1 and CB2), thus providing unprecedented evidence of ECS dysregulation in RPE cells upon BL exposure.
Collapse
Affiliation(s)
- Camilla Di Meo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy; Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Annamaria Tisi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Anna Rita Lizzi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Sakthimala Palaniappan
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Fanny Pulcini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Marc Nazarè
- Leibniz Research Institute for Molecular Pharmacology (FMP), Campus Berlin-Buch, 13125, Berlin, Germany
| | - Eric Hsu
- InMed Pharmaceuticals Inc., Vancouver, BC, Canada
| | - Cinzia Rapino
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy; European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, Rome, Italy.
| |
Collapse
|
107
|
Xu M, Zhang QT, Zhou L, Cai YW, Liu H, Zhao QL, Tian JH, Huang YK, Ren P, Huang X. Ferulic acid in Chaihu Shugan San modulates depression-like behavior, endothelial and gastrointestinal dysfunction in rats via the Ghrl-Edn1/Mecp2/P-mTOR/VEGFA pathway: A multi-omics study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119624. [PMID: 40127829 DOI: 10.1016/j.jep.2025.119624] [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: 11/15/2024] [Revised: 01/16/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In a global context of escalating multimorbidity, characterized by the co-occurrence of major depressive disorder (MDD), endothelial dysfunction (ED), and gastrointestinal dysregulation (GD), the quest for effective treatments has become paramount. Central to these interconnected conditions is oxidative stress (OS), a pivotal factor that has been extensively studied yet remains inadequately addressed. This study introduces Chaihu-Shugan-San (CSS) and its absorbed component ferulic acid (FA), a potent antioxidant derived from medicinal plants, as a novel therapeutic approach with the unique ability to counter the multifaceted effects of acute forced swimming (AFS)-induced depression, ED, and GD. Unlike traditional single-disease-focused studies, our research explores the synergistic effects of CSS and FA across these interrelated disorders, offering a groundbreaking perspective. AIM OF THE STUDY This study aims to evaluate CSS and FA in treating depression-related multimorbidity triggered by AFS and to uncover the shared underlying mechanisms of FA. MATERIALS AND METHODS A depression-like model in rats was induced by AFS, and an OS model was established in endothelial cells (ECs) through hydrogen peroxide treatment. We investigated the effects of CSS and FA on MDD, ED, and GD in rats and OS levels in ECs. Our assessments included hematoxylin and eosin (HE) staining, biochemical assays, and behavioral studies. We conducted an integrated analysis of transcriptomics, proteomics, and phosphoproteomics data to elucidate the underlying mechanisms. The identification of relevant targets was confirmed through Western blotting (WB), real-time quantitative polymerase chain reaction (RT-qPCR), molecular docking studies, and an extensive literature review. RESULTS Our findings indicate that CSS and FA not only significantly mitigate AFS-induced abnormalities in the open field test (OFT), forced swim test (FST), and related behaviors such as gastric emptying and intestinal transit in rats but also ameliorate depression, ED, GD, inflammation and OS-related biomarker levels, alongside HE staining in gastric sinus and aorta slices. The study also highlights that FA can influence OS and endothelial function in ECs. Moreover, a combined multi-omics analysis unveiled several OS-related pathways, including the mTOR and p53 signaling pathways. Our research elucidates that the Ghrl-Edn1/Mecp2/P-mTOR/Vegfa-associated OS signaling pathway is pivotal in countering AFS-induced multimorbidity, expanding beyond the conventional disease-specific focus. CONCLUSIONS This pioneering study underscores capability of CSS and FA to tackle AFS-induced multimorbidity concurrently and intricately details FA's antioxidative mechanisms within ECs. The insights gleaned offer a novel perspective on FA's role in multimorbidity regulation and its potential to modulate OS, especially in the complex environment of ECs. Given the urgent global health challenges, our research positions FA as a promising therapeutic contender, advocating for a paradigm shift in multimorbidity management.
Collapse
Affiliation(s)
- Min Xu
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qian-Tao Zhang
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Zhou
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ya-Wen Cai
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Liu
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiu-Long Zhao
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-Hua Tian
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun-Ke Huang
- Women's Hospital, Zhejiang University School of Medicine, Gynecology Department, Zhejiang, China
| | - Ping Ren
- Department of Geriatrics, Jiangsu Province Hospital of TCM, Nanjing, China
| | - Xi Huang
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
108
|
Jin Y, Liu J, Zhang X, Zhang L, Cui Y, Luo X, Zhu H, Chen Z, Liu M, Wu X, Chen X, Liao S, Wu G, Fang X, Meng Q. Stage-dependent proteomic alterations in aqueous humor of diabetic retinopathy patients based on data-independent acquisition and parallel reaction monitoring. J Transl Med 2025; 23:476. [PMID: 40281624 PMCID: PMC12032686 DOI: 10.1186/s12967-025-06452-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Diabetic retinopathy (DR), a microvascular complication of diabetes mellitus (DM), represents the predominant cause of preventable vision loss in working-age populations globally. While the pathophysiological mechanisms underlying DR progression remain incompletely understood, our study employs comprehensive proteomic profiling of aqueous humor (AH) to identify stage-specific biomarkers and therapeutic targets in type 2 diabetes mellitus (T2DM) patients across DR progression. METHODS Utilizing data-independent acquisition (DIA) mass spectrometry, we quantified AH proteomes in a discovery cohort comprising 24 subjects: 18 T2DM patients stratified by DR severity [6 non-DR, 6 non-proliferative DR (NPDR), 6 proliferative DR (PDR)] and 6 cataract controls without diabetes (non-DM). Validation cohort analysis (including 10 AH samples in each group) was performed using parallel reaction monitoring (PRM) strategy for verification of target proteins. Comprehensive bioinformatics analyses included gene set enrichment analysis (GSEA), weighted gene co-expression network analysis (WGCNA), Kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis, protein-protein interaction (PPI) network construction, receiver operating characteristic (ROC) curve analysis, and ConnectivityMap (Cmap)-based drug prediction. RESULTS Proteomic profiling identified 739 quantifiable AH proteins (62% extracellular) with clear separation among the four clinical stages in the discovery cohort. GSEA uncovered altered expression of proteins mainly related to complement and coagulation cascades, folate metabolism, and the selenium micronutrient network in patients with DR. WGCNA-derived protein modules yielded 83 PRM-validated targets, including 5 hub proteins differentiating NPDR from non-DR and 33 hub proteins showed significant upregulation in PDR versus NPDR comparison. Clinical correlation analysis identified F2, FGG, FGB, RBP4, AMBP, VTN, C8A, CPB2, and C2 associated with clinical traits. C6, FAM3C, SPP1, and JCHAIN levels were altered post-anti-VEGF treatment. Pharmacological prediction identified potential therapeutic compounds, including perindopril, triciribine, and XAV-939 for NPDR, and topiramate, triciribine, and vecuronium for PDR. CONCLUSION This study established a comprehensive AH proteomic signature of DR progression, offering insights into the pathogenesis of DR and highlighting potential biomarkers and novel therapeutic targets.
Collapse
Affiliation(s)
- Yeanqi Jin
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Junbin Liu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xueli Zhang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Liang Zhang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ying Cui
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiaoyang Luo
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Haoxian Zhu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhifan Chen
- The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mengya Liu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiyu Wu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xinyu Chen
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shuoxin Liao
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guanrong Wu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiang Fang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Qianli Meng
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
109
|
Li YL, Li Y, Tu H, Evans AJ, Patel TA, Zheng H, Patel KP. Stellate Ganglia: A Key Therapeutic Target for Malignant Ventricular Arrhythmia in Heart Disease. Circ Res 2025; 136:1049-1069. [PMID: 40273204 PMCID: PMC12026290 DOI: 10.1161/circresaha.124.325384] [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] [Indexed: 04/26/2025]
Abstract
Malignant ventricular arrhythmias (VAs), such as ventricular tachycardia and ventricular fibrillation, are the cause of approximately half a million deaths per year in the United States, which is a common lethal event in heart disease, such as hypertension, catecholaminergic polymorphic ventricular tachycardia, takotsubo cardiomyopathy, long-QT syndrome, and progressing into advanced heart failure. A common characteristic of these heart diseases, and the subsequent development of VAs, is the overactivation of the sympathetic nervous system. Current treatments for VAs in these heart diseases, such as β-adrenergic receptor blockers and cardiac sympathetic ablation, aim at inhibiting cardiac sympathetic overactivation. However, these treatments do not translate into becoming efficacious as long-term suppressors of ventricular tachycardia/ventricular fibrillation events. As a key regulatory component in the heart, cardiac postganglionic sympathetic neurons residing in the stellate ganglia (SGs) release neurotransmitters (such as norepinephrine and NPY [neuropeptide Y]) to perform their regulatory role in dictating cardiac function. Growing evidence from animal experiments and clinical studies has demonstrated that the remodeling of the SG may be intimately involved in malignant arrhythmogenesis. This identifies the SG as a key potential therapeutic target for the treatment of malignant VAs in heart disease. Therefore, this review summarizes the role of SG in ventricular arrhythmogenesis and updates the novel targeting of SG for clinical treatment of VAs in heart disease.
Collapse
Affiliation(s)
- Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yu Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Huiyin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anthony J. Evans
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tapan A. Patel
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hong Zheng
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Kaushik P. Patel
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
110
|
Paloczi J, Gunduz-Cinar O, Yokus B, Paes-Leme B, Haskó G, Kunos G, Holmes A, Pacher P. Exacerbated cardiac dysfunction from combined alcohol binge and synthetic cannabinoid use. Biomed Pharmacother 2025; 187:118053. [PMID: 40288176 DOI: 10.1016/j.biopha.2025.118053] [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: 07/08/2024] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Alcohol remains the most frequently used intoxicant, posing a significant global health concern. Binge drinking has been linked to acute cardiovascular complications, including reduced cardiac performance, arrhythmias, and blood pressure instability. Additionally, there is a growing number of clinical reports describing severe adverse cardiac events associated with the recreational use of synthetic cannabinoids. Recent surveys reveal a troubling rise in polydrug misuse, particularly among young adults, with an increasing number of cases linked to fatal outcomes. This study aimed to characterize left ventricular performance in mice following combined acute alcohol and synthetic cannabinoid exposure using complex hemodynamic measurements via the pressure-volume (P-V) approach. Our findings revealed that alcohol ingestion or intravenous synthetic cannabinoid (CP55,940) administration led to a dose-dependent decline in systolic cardiac performance in mice. Moreover, the concurrent administration of alcohol and CP55,940 led to cardiodepression, surpassing the contractile dysfunction observed with each drug administered individually. Intravenous administration of the cannabinoid type-1 receptor (CB1R) antagonist rimonabant largely improved the combined drug administration-induced left ventricular contractile dysfunction in mice, while its intracerebroventricular administration resulted in only partial restoration of normal cardiac function, implicating a role for both central and peripheral CB1R signaling. Our results emphasize the severe cardiac consequences of simultaneous alcohol and synthetic cannabinoid misuse and offer a potential therapeutic avenue for mitigating the adverse cardiac effects of their combined use by repurposing CB1R antagonists.
Collapse
Affiliation(s)
- Janos Paloczi
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD 20852, USA; Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Ozge Gunduz-Cinar
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse, NIH, Bethesda, MD, USA
| | - Burhan Yokus
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD 20852, USA
| | - Bruno Paes-Leme
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD 20852, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY 10032, USA
| | - George Kunos
- Laboratory of Physiological Studies, National Institutes of Health/NIAAA, Bethesda, MD 20852, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse, NIH, Bethesda, MD, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD 20852, USA.
| |
Collapse
|
111
|
Zhou W, Zhou L, Qi Z, Dai S, Zhang P, Zhong H, Xu H, Zhao X, Lian X, Lin J, Wu H. The soluble guanylate cyclase (sGC) stimulator vericiguat inhibits platelet activation and thrombosis. Eur J Pharmacol 2025; 999:177670. [PMID: 40287044 DOI: 10.1016/j.ejphar.2025.177670] [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: 12/10/2024] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Vericiguat, a soluble guanylate cyclase (sGC) stimulator, is used to treat chronic heart failure. Vericiguat can directly bind to the sGC in the absence of NO or stabilize the binding of NO to sGC, thereby stimulating cGMP production. Vericiguat correlates closely with the platelet activation. However, the precise effect of vericiguat on platelet activation and thrombosis in vivo remains to be elucidated. METHODS We investigated the effects of vericiguat on agonist-induced platelet aggregation, secretion, integrin αIIbβ3 activation, spreading, clot retraction, and thrombus formation in vivo, elucidating the underlying mechanisms. Additionally, we performed whole blood aggregometry and Microfluidic whole-blood perfusion assay to determine whether vericiguat could alleviate thrombosis. RESULTS Vericiguat concentration-dependently inhibited aggregation and ATP release induced by agonists both in human and mouse platelets. P-selection expression, integrin αIIbβ3 activation, spreading, and clot retraction induced by thrombin were all inhibited by vericiguat. Mechanistically, vericiguat bound to the sGC in platelets, avtivating the cGMP/PKG signaling pathway to inhibit the platelet. Vericiguat also inhibited the FeCl3-injured thrombus formation in mesenteric arterioles in wild-type (WT) mice and pulmonary vascular thrombi after constructing the pulmonary embolism model. Oral administration of vericiguat for 2 weeks attenuated thromboembolism in brain, too. CONCLUSION Vericiguat directly inhibits platelet activation and thrombosis in vivo by binding to the sGC and activating cGMP/PKG pathway. In addition to the treatment for chronic heart failure, it may have therapeutic advantages in treating thrombotic diseases.
Collapse
Affiliation(s)
- Wenxuan Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Luning Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Zhiyong Qi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Shimo Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Peng Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Haoxuan Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Huajie Xu
- Department of Infectious Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Xiaoyu Lian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China
| | - Jiaxiong Lin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China.
| | - Hongyi Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, China.
| |
Collapse
|
112
|
Mirani B, Dauz JD, Yazaki K, Latifi N, Santerre JP, Bendeck MP, Simmons CA, Friedberg MK. Right Ventricular Stiffening and Function Are Associated With Main Pulmonary Artery Remodeling in a Rat Model of Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2025. [PMID: 40270258 DOI: 10.1161/atvbaha.124.321354] [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: 06/06/2024] [Accepted: 03/26/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Coupling between right ventricular (RV) function and the pulmonary vasculature determines outcomes in pulmonary arterial (PA) hypertension. The mechanics of the main PA (mPA) is an important but understudied determinant of RV-PA coupling. To investigate the histology and mechanics of PA in relationship to RV remodeling, mechanics, hemodynamics, and coupling in experimental PA hypertension. METHODS In a sugen-hypoxia rat model of PA hypertension, RV hemodynamics were assessed by conductance catheters. Active tension-strain curves were generated using echocardiography. mPA and RV free wall were harvested to determine their macrostructure and microstructure, composition, and mechanical properties. Comprehensive multivariate analyses elucidated relationships between PA and RV mechanics, structure, and coupling. RESULTS Pulmonary hypertensive mPAs developed fibrosis relative to healthy controls, as did RVs, which also hypertrophied, with reorientation of muscle fibers toward a trilayer architecture reminiscent of normal left ventricular architecture. Increased glycosaminoglycan deposition and increased collagen-to-elastin ratio in PA, and increased collagen, as well as hypertrophy and reorganization of myofibers in RV, led to increased stiffness. This increase in stiffness was more pronounced in the longitudinal direction in the high- and low-strain regime for PA and RV, respectively, causing increased mechanical anisotropy. mPA stiffening correlated significantly with RV tissue mechanical remodeling and reduced systolic performance, cardiac output, and RV-PA coupling. CONCLUSIONS Compositional, structural, and mechanical changes in mPA correlate with adverse RV remodeling, mechanics, function, and coupling in PA hypertension. Therefore, increasing mechanical compliance of the large PAs may be an important and novel therapeutic strategy for mitigating RV failure.
Collapse
Affiliation(s)
- Bahram Mirani
- Department of Mechanical and Industrial Engineering, University of Toronto, ON, Canada. (B.M., N.L., C.A.S.)
- Institute of Biomedical Engineering, University of Toronto, ON, Canada. (B.M., J.P.S., C.A.S.)
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada (B.M., N.L., J.P.S., M.P.B., C.A.S.)
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (B.M., K.Y.)
| | - John D Dauz
- Labatt Family Heart Center, The Hospital for Sick Children, University of Toronto, ON, Canada. (J.D.D., M.K.F.)
- Department of Paediatrics, University of Toronto, ON, Canada. (J.D.D., M.K.F.)
| | - Kana Yazaki
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (B.M., K.Y.)
| | - Neda Latifi
- Department of Mechanical and Industrial Engineering, University of Toronto, ON, Canada. (B.M., N.L., C.A.S.)
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada (B.M., N.L., J.P.S., M.P.B., C.A.S.)
- Department of Medical Engineering, University of South Florida, Tampa (N.L.)
| | - J Paul Santerre
- Institute of Biomedical Engineering, University of Toronto, ON, Canada. (B.M., J.P.S., C.A.S.)
- Faculty of Dentistry, University of Toronto, ON, Canada. (J.P.S.)
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada (B.M., N.L., J.P.S., M.P.B., C.A.S.)
| | - Michelle P Bendeck
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada. (M.P.B.)
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada (B.M., N.L., J.P.S., M.P.B., C.A.S.)
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, ON, Canada. (B.M., N.L., C.A.S.)
- Institute of Biomedical Engineering, University of Toronto, ON, Canada. (B.M., J.P.S., C.A.S.)
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada (B.M., N.L., J.P.S., M.P.B., C.A.S.)
| | - Mark K Friedberg
- Labatt Family Heart Center, The Hospital for Sick Children, University of Toronto, ON, Canada. (J.D.D., M.K.F.)
- Department of Paediatrics, University of Toronto, ON, Canada. (J.D.D., M.K.F.)
| |
Collapse
|
113
|
Zhao Y, Xu T, Wu Z, Li N, Liang Q. Rebalancing redox homeostasis: A pivotal regulator of the cGAS-STING pathway in autoimmune diseases. Autoimmun Rev 2025; 24:103823. [PMID: 40286888 DOI: 10.1016/j.autrev.2025.103823] [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: 07/11/2024] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Autoimmune diseases (ADs) arise from the breakdown of immune tolerance to self-antigens, leading to pathological tissue damage. Proinflammatory cytokine overproduction disrupts redox homeostasis across diverse cell populations, generating oxidative stress that induces DNA damage through multiple mechanisms. Oxidative stress-induced alterations in membrane permeability and DNA damage can lead to the recognition of double-stranded DNA (dsDNA), mitochondrial DNA (mtDNA) and micronuclei-DNA (MN-DNA) by DNA sensors, thereby initiating activation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. While previous reviews have characterized cGAS-STING activation in autoimmunity, the reciprocal regulation between redox homeostasis and cGAS-STING activation remains insufficiently defined. This narrative review examines oxidative stress-mediated DNA damage as a critical driver of pathological cGAS-STING signaling and delineates molecular mechanisms linking redox homeostasis to autoimmune pathogenesis. Furthermore, we propose therapeutic strategies that combine redox restoration with the attenuation of aberrant cGAS-STING activation, thereby establishing a mechanistic foundation for precision interventions in autoimmune disorders. METHODS: The manuscript is formatted as a narrative review. We conducted a comprehensive search strategy using electronic databases such as PubMed, Google Scholar and Web of Science. Various keywords were used, such as "cGAS-STING," "Redox homeostasis," "Oxidative stress," "pentose phosphate pathway," "Ferroptosis," "mtDNA," "dsDNA," "DNA damage," "Micronuclei," "Reactive oxygen species," "Reactive nitrogen species," "Nanomaterial," "Autoimmune disease," "Systemic lupus erythematosus," "Type 1 diabetes," "Rheumatoid arthritis," "Multiple sclerosis," "Experimental autoimmune encephalomyelitis," "Psoriasis," etc. The titles and abstracts were reviewed for inclusion into this review. After removing duplicates and irrelevant studies, 174 articles met inclusion criteria (original research, English language).
Collapse
Affiliation(s)
- Yuchen Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Tianhao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China
| | - Zhaoshun Wu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China
| | - Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China.
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
114
|
Teng X, Wang Q, Ma J, Li D. Integrating bioinformatics and machine learning to discover sumoylation associated signatures in sepsis. Sci Rep 2025; 15:14398. [PMID: 40274894 PMCID: PMC12022290 DOI: 10.1038/s41598-025-96956-x] [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: 10/09/2024] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Small Ubiquitin-like MOdifier-mediated modification (SUMOylation) is associated with sepsis; however, its molecular mechanism remains unclear. Herein, hub genes and regulatory mechanisms in sepsis was investigated. The GSE65682 and GSE95233 datasets were extracted from public databases. Differential analysis and Weighted Gene Co-expression Network Analysis (WGCNA) were conducted in GSE65682 to identify differentially expressed genes (DEGs) and key module genes. Candidate genes were derived by intersecting with SUMOylation-related genes (SUMO-RGs). The Least Absolute Shrinkage and Selection Operator (LASSO) and Support Vector Machine-Recursive Feature Elimination (SVM-RFE) were utilized to identify significant feature genes. The convergence of those genes was utilized for diagnostic assessment and expression validation. Hub genes were defined as those exhibiting an area under the curve (AUC) greater than 0.7, significant gene expression, and a consistent trend. Localization and functional analyses of hub genes were conducted to enhance the understanding of these genes. Immune analysis, regulatory network construction, and drug prediction were performed. Six hub genes were identified: RORA, L3MBTL2, PHC1, RPA1, CHD3, and RANGAP1. These genes possessed considerable diagnostic significance for sepsis and were also markedly downregulated in the condition. Hub genes were predominantly enriched in the ribosome pathway and exhibited a strong correlation with differential immune cells. Activated CD8 + T cells exhibited a positive correlation with RORA. Based on the predicted and established regulatory network, AC004687.1 was observed to modulate PHC1 expression via hsa-miR- 142 - 5p. A total of six hub genes (RORA, L3MBTL2, PHC1, RPA1, CHD3, and RANGAP1) associated with SUMOylation was identified in sepsis in the current study. The findings are likely to aid in the differentiation between control and disease states, offering substantiation for the diagnosis of sepsis.
Collapse
Affiliation(s)
- Xue Teng
- Department of Anesthesiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, Heilongjiang, China
| | - Qi Wang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jinling Ma
- Department of Intensive Care Medicine, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Dongmei Li
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, Heilongjiang, China.
| |
Collapse
|
115
|
Almohaimeed GM, Alonazi AS, Alshammari TK, Bin Dayel AF, Alghibiwi HK, Alamin MA, Almotairi AR, Aldawsari NA, Alkhelb DA, Alrasheed NM, Sarawi WS, Alrasheed NM. Metformin-mediated protection against Immunosenescence in diabetic cardiomyopathy: The potential roles of GDF-15 and klotho proteins. Int Immunopharmacol 2025; 153:114530. [PMID: 40139098 DOI: 10.1016/j.intimp.2025.114530] [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: 10/03/2024] [Revised: 03/07/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a global health concern. However, studies examining the effect of metformin on diabetes-induced cardiac myocyte aging are lacking. This study aimed to investigate the protective effect of metformin against DCM involving modulation of macrophage phenotypes, growth differentiation factor-15 (GDF-15), and the anti-aging protein Klotho. Diabetes was induced in male Wistar rats using streptozotocin. Diabetic and nondiabetic rats were treated with metformin (200 mg/kg/day) and saline (control). DCM, inflammation, adhesion molecules, immunometabolic, and GDF-15 biomarkers were assessed using immunoassays. Western blotting was used to analyze Klotho expression. Macrophage phenotypes, senescence-associated-galactosidase (SA-β-gal), and p16INK4a were examined using immunohistochemistry, whereas the heart sections were histologically examined. The untreated diabetic rats showed increased serum troponin I and creatine kinase-MB levels, reflecting cardiac damage, which was confirmed via morphological changes and senescence. Klotho expression was decreased, indicating cardiac aging. Treatment with metformin reduced the heart weight-body weight ratio and lowered cardiac injury, inflammation, and adhesion molecule biomarker levels. It also reversed the histopathological changes induced by diabetes. It shifted macrophage polarization toward the M2 phenotype, decreased p16INK4a and SA-β-gal expression, and enhanced Klotho and GDF-15 expression. These findings revealed that diabetes induces cardiac aging by increasing senescence markers and decreasing the expression of Klotho. Metformin treatment protects against DCM by modulating macrophage phenotypes, attenuating immunosenescence-related dysregulation, and enhancing GDF-15 and Klotho expressions. Thus, metformin has potential clinical implications in alleviating DCM.
Collapse
Affiliation(s)
- Ghada M Almohaimeed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Asma S Alonazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Tahani K Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Anfal F Bin Dayel
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hanan K Alghibiwi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Maha A Alamin
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmad R Almotairi
- Department of Pathology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Nasser A Aldawsari
- Pathology Department, Security Forces Hospital, Riyadh 11564, Saudi Arabia
| | - Dalal A Alkhelb
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nawal M Alrasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Wedad S Sarawi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nouf M Alrasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
116
|
Shokrzadeh S, Moghim S, Shokrzadeh M, Aghajanshakeri S. An added value of azithromycin: mitigation of doxorubicin-associated oxidative damage and genotoxicity in normal human bronchial epithelium cells. Mutagenesis 2025; 40:126-136. [PMID: 39373202 DOI: 10.1093/mutage/geae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/05/2024] [Indexed: 10/08/2024] Open
Abstract
Doxorubicin, a well-known and widely used antineoplastic agent with direct ROS-accumulating activity, has proven effective in treating various cancer types. However, its non-specific cytotoxicity towards non-cancerous cells prompts concerns regarding potential adverse effects. Azithromycin is an antibiotic for treating bacterial infections and an anti-inflammatory agent, particularly beneficial in managing respiratory conditions like bronchitis and sinusitis. Despite azithromycin's well-documented antibacterial properties, its potential cellular/genomic protective effects remain unexplored. As an in vitro model, BEAS-2B cells (normal human bronchial epithelium cells) were employed in this study to assess whether azithromycin possesses any protective properties against doxorubicin-induced cellular toxicity. Cells in pretreatment culture were treated to various amounts of azithromycin (3.125, 6.25, 12.5, 25, and 50 μg/ml) in combination with doxorubicin at IC50 (0.08 μg/ml). Doxorubicin at 0.08 μg/ml highlighted cytotoxicity, oxidative stress, and genotoxicity. Azithromycin at 25 and 50 μg/ml markedly modulated oxidative stress and genomic damage by decreasing the ROS and LPO amounts and suppressing DNA fragmentation in the comet assay parameters. Consequently, azithromycin may be regarded as a cytomodulating, antigenotoxic, and antioxidant agent.
Collapse
Affiliation(s)
| | - Shahrzad Moghim
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Shokrzadeh
- Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shaghayegh Aghajanshakeri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
117
|
Ke A, Yang W, Zhang W, Chen Y, Meng X, Liu J, Dai D. The cardiac glycoside periplocymarin sensitizes gastric cancer to ferroptosis via the ATP1A1-Src-YAP/TAZ-TFRC axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156804. [PMID: 40311597 DOI: 10.1016/j.phymed.2025.156804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Targeting ferroptosis vulnerabilities in tumors has become an increasingly promising therapeutic strategy. While the regulatory effects of natural products on ferroptosis are progressively being elucidated, the role of cardiac glycosides in modulating ferroptosis remains poorly understood. PURPOSE This study aims to investigate the ferroptosis-sensitizing effects of periplocymarin (PPM), a cardiac glycoside derived from the traditional plant Periploca sepium, and to elucidate the underlying molecular mechanisms. METHODS The effects of PPM on ferroptosis regulation were comprehensively assessed through functional assays, followed by sequencing analysis to identify associated signaling pathways. Subsequent mechanistic validation experiments were conducted to confirm the upstream and downstream regulatory components involved in this ferroptosis-modulating axis. RESULTS PPM induced slow and mild apoptosis in gastric cancer cells through the inhibition of glycolysis. However, when combined with ferroptosis inducers, it promoted rapid and robust ferroptosis. In vivo, PPM sensitized gastric cancer xenografts to cisplatin-induced ferroptosis with no observable cardiotoxicity or renal impairment. Mechanistically, PPM targeted the α1 subunit of the Na+/K+-ATPase (ATP1A1), leading to the activation of Src, which subsequently induced tyrosine phosphorylation of YAP/TAZ in a Hippo-independent manner, promoting their nuclear translocation. The YAP/TAZ-TEAD transcriptional complex directly bound to the TFRC promoter region between nucleotides 401-409 upstream of the transcription start site, thereby activating TFRC transcription. This resulted in increased iron influx, elevated lipid peroxidation, and heightened sensitivity to ferroptosis. Notably, ATP1A1 was essential for ferroptosis resistance, as its knockdown mimicked the sensitizing effect of PPM on ferroptosis. Moreover, the oncogenic Src-YAP/TAZ-TFRC axis may have represented a ferroptosis vulnerability and a potential biomarker in ferroptosis therapy for cancer. Importantly, other cardiac glycosides targeting Na+/K+-ATPase, such as digitoxin and bufalin, also enhanced ferroptosis sensitivity in gastric cancer cells through activation of YAP/TAZ signaling. CONCLUSION Our findings establish the cardiac glycoside PPM as a novel ferroptosis sensitizer that targets ATP1A1 to activate the Src-YAP/TAZ-TFRC axis, providing mechanistic insights for repurposing cardiac glycosides as ferroptosis modulators in precision combinatorial cancer therapy.
Collapse
Affiliation(s)
- Angting Ke
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Weiguang Yang
- Department of Nephrology, the First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Wanchuan Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yibin Chen
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xiangyu Meng
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China
| | - Jie Liu
- Translational Research Experiment Department, Science Experiment Center, China Medical University, Shenyang 110122, China
| | - Dongqiu Dai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
118
|
Ronnacker J, Grau M, Klasmeier M, Klesse C, Baldauf H, Abert S, Berdel AF, Füsser FT, Sandmann S, Albring J, Reicherts C, Call S, Marx J, Floeth M, Eßeling E, Kolloch L, Berning P, Scheller A, Wethmar K, Schmidt H, Schlüter B, Berdel WE, Ostendorf BN, Tavazoie SF, Mikesch JH, Lenz G, Fleischhauer K, Schetelig J, Stelljes M, Schliemann C. Common hereditary variants of the APOE gene and posttransplant outcome in acute myeloid leukemia. Blood 2025; 145:1943-1956. [PMID: 39813679 DOI: 10.1182/blood.2024026886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/23/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025] Open
Abstract
ABSTRACT Apolipoprotein E (APOE) has multiple functions in metabolism and immunoregulation. Its common germ line variants APOE2, APOE3, and APOE4 give rise to 3 functionally distinct gene products. Previous studies reported yin-yang roles of APOE2 and APOE4 in immunological processes, but their effects in hematopoietic stem cell transplantation (HSCT) have never been studied. We performed APOE genotyping in 2 contemporary cohorts of 348 and 447 patients with acute myeloid leukemia who had received allogeneic HSCT and evaluated the associations of recipient and donor APOE genetic variations with posttransplant outcomes. Patients who carried at least 1 APOE2 allele had a higher risk of posttransplant death than APOE4 carriers in the discovery (hazard ratio [HR], 2.09; P = .024) and validation cohorts (HR, 1.96; P = .040). Detrimental APOE2 effects were driven by an increased risk of severe chronic graft-versus-host disease (GVHD; adjusted HR [HRadj], 1.85; P = .034) and nonrelapse death (HRadj, 1.72; P = .044). In non-APOE2 recipients, transplantation of an APOE2-positive allograft was associated with an increased incidence of grade 3 to 4 acute GVHD (HRadj, 2.82; P = .012) and severe chronic GVHD (HRadj, 2.54; P = .022) compared with APOE2-negative grafts. In summary, the APOE2 allele, typically considered a longevity gene in the general population, was associated with a higher risk of acute GVHD (HRadj, 2.75; P = .002), chronic GVHD (HRadj, 2.57; P = .001), and posttransplant mortality (HRadj, 1.79; P = .004), when present in either the host or transplanted from the donor.
Collapse
Affiliation(s)
- Julian Ronnacker
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Michael Grau
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | | | | | | | - Stefan Abert
- Collaborative Biobank, DKMS Group gGmbH, Tübingen, Germany
| | - Andrew F Berdel
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Friederike T Füsser
- Institute for Pharmaceutical and Medical Chemistry, University of Münster, Münster, Germany
| | - Sarah Sandmann
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Jörn Albring
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | | | - Simon Call
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Julia Marx
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Matthias Floeth
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Eva Eßeling
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Lina Kolloch
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Philipp Berning
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Annika Scheller
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Klaus Wethmar
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Hartmut Schmidt
- Center for Laboratory Medicine, University Hospital Münster, Münster, Germany
| | - Bernhard Schlüter
- Center for Laboratory Medicine, University Hospital Münster, Münster, Germany
| | - Wolfgang E Berdel
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Benjamin N Ostendorf
- Department of Hematology, Oncology, and Tumor Immunology, Berlin Institute of Health, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sohail F Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY
| | | | - Georg Lenz
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | | | - Johannes Schetelig
- Collaborative Biobank, DKMS Group gGmbH, Tübingen, Germany
- Department of Medicine I, University Hospital Dresden, Dresden, Germany
| | - Matthias Stelljes
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | | |
Collapse
|
119
|
Mikdar M, Serra M, Azouzi S. Adenosine signaling in promoting the balance between erythropoiesis and myelopoiesis. Curr Opin Hematol 2025:00062752-990000000-00110. [PMID: 40314242 DOI: 10.1097/moh.0000000000000872] [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: 05/03/2025]
Abstract
PURPOSE OF REVIEW Adenosine signaling is emerging as a key regulator of hematopoietic lineage commitment, influencing both erythropoiesis and myelopoiesis. This review explores the distinct roles of adenosine receptors in balancing these processes, particularly under stress conditions. Since adenosine extracellular levels are increased in multiple hematological disorders, including sickle cell disease, deciphering the mechanisms downstream of adenosine receptor activation is crucial to understand the pathophysiology of these conditions. RECENT FINDINGS Extracellular adenosine levels in the bone marrow microenvironment are tightly regulated by CD39/CD73 activity and ENT1 uptake. Recent studies have shown that ENT1-mediated adenosine transport is crucial for adenosine intracellular metabolism and normal erythropoiesis, while increased extracellular adenosine levels impact hematopoietic differentiation through adenosine receptor activation.. High dose of exogenous adenosine inhibits erythroid proliferation by inducing G1 arrest and p53-mediated apoptosis. Furthermore, A2B and A3 receptor signaling inhibits erythroid differentiation, while adenosine signaling through A3 also favors granulopoiesis. SUMMARY Collectively, these findings highlight adenosine signaling as a critical and multifaceted regulator of hematopoietic balance, offering novel insights into its therapeutic potential for managing disorders characterized by ineffective erythropoiesis and aberrant myelopoiesis.
Collapse
Affiliation(s)
- Mahmoud Mikdar
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Marion Serra
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Slim Azouzi
- Université Paris Cité and Université des Antilles, INSERM, EFS, UMR S1134, Laboratory of Blood Group Antigens, Hematopoiesis and Sickle Cell Disease, Paris, France
| |
Collapse
|
120
|
Zhou ZY, Zhou ZP, Yue YX, Zhong YK, Yang ZX, Cai GL. Dimethyl Pent-2-Enedioate inhibits LPS-induced inflammatory response in macrophages. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025:S1684-1182(25)00084-2. [PMID: 40300966 DOI: 10.1016/j.jmii.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/01/2025] [Accepted: 03/27/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Endogenous metabolite itaconate and its derivative Dimethyl itaconate (DMI) exhibit significant anti-inflammatory effects. Dimethyl Pent-2-Enedioate (DMP), an isomer of DMI, may possess similar properties. This study investigates the anti-inflammatory effects of DMP in LPS-induced macrophages and explores its potential regulatory mechanisms. METHODS Inflammatory marker levels were assessed at both the mRNA and protein levels using ELISA and qRT-PCR. The activation status of macrophages was evaluated by flow cytometry, quantifying the number of CD40-positive cells. RNA sequencing was conducted to investigate the transcriptomic changes following DMP treatment. Subsequent GO and KEGG enrichment analyses were performed to identify potential mechanisms underlying DMP's effects. Western blot analysis was employed to assess the expression of p-p65, while immunofluorescence analysis was used to examine p65 nuclear translocation, providing insight into the regulatory effects of DMP on the NF-κB signaling pathway. RESULTS DMP inhibited the expression of inflammatory markers TNF-α, IL-6, and MCP-1 at both mRNA and protein levels. Flow cytometry analysis revealed a decrease in CD40-positive cells. RNA sequencing identified DEGs enriched in inflammation-related pathways. Western blotting and immunofluorescence confirmed that DMP reduced p-p65 expression and inhibited p65 nuclear translocation, suggesting a potential regulatory effect on the NF-κB signaling pathway. CONCLUSION DMP significantly inhibits LPS-induced inflammation in macrophages, with its underlying mechanisms being complex. Our data demonstrate that DMP exerts its anti-inflammatory effects at least in part through the downregulation of the NF-κB pathway, offering potential applications in the prevention and treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Zhi-Ying Zhou
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhi-Peng Zhou
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying-Xing Yue
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yu-Ke Zhong
- Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhou-Xin Yang
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.
| | - Guo-Long Cai
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
121
|
Bhattacharyya A, Vasconcelos D, Spicarova D, Palecek J. 20:4-NAPE induced changes of mechanical sensitivity and DRG neurons excitability are concentration dependent and mediated via NAPE-PLD. Sci Rep 2025; 15:14131. [PMID: 40269193 PMCID: PMC12019079 DOI: 10.1038/s41598-025-98567-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
Alterations in the excitability of dorsal root ganglion (DRG) neurons are critical in the pathogenesis of acute and chronic pain. Neurotransmitter release from the terminals of DRG neurons is regulated by cannabinoid receptor 1 (CB1) and transient receptor potential vanilloid 1 (TRPV1), both activated by anandamide (AEA). In our experiments, the AEA precursor N-arachidonoylphosphatidylethanolamine (20:4-NAPE) was used to study the modulation of nociceptive DRG neurons excitability using K+-evoked Ca2+ transients. Intrathecal administration was used to evaluate in vivo effects. Application of 20:4-NAPE at lower concentrations (10 nM - 1 µM) decreased the excitability of DRG neurons, whereas the higher (10 µM) increased it. Both effects of 20:4-NAPE were blocked by the N-acylphosphatidylethanolamine phospholipase D (NAPE-PLD) inhibitor LEI-401. Similarly, lower concentrations of externally applied AEA (1 nM - 10 nM) inhibited DRG neurons, whereas higher concentration (100 nM) did not change it. High AEA concentration (10 µM) evoked Ca2+ transients dependent on TRPV1 activation in separate experiments. Inhibition of the CB1 receptor by PF514273 (400 nM) prevented the 20:4-NAPE- and AEA-induced inhibition, whereas TRPV1 inhibition by SB366791 (1 µM) prevented the increased DRG neuron excitability. In behavioral tests, lower 20:4-NAPE concentration caused hyposensitivity, while higher evoked mechanical allodynia. Intrathecal LEI-401 prevented both in vivo effects of 20:4-NAPE. These results highlight anti- and pro-nociceptive effects of 20:4-NAPE mediated by CB1 and TRPV1 in concentration-dependent manner. Our study underscores the complexity of endocannabinoid signaling in pain transmission modulation and highlights 20:4-NAPE as a potential therapeutic target, offering new insights for developing analgesic strategies.
Collapse
Affiliation(s)
- Anirban Bhattacharyya
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Daniel Vasconcelos
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Diana Spicarova
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Palecek
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
122
|
Ledderose C, Valsami EA, Elevado M, Stevenson A, Abutabikh R, Curatolo J, Junger WG. Adenosine accumulation in the blood of newborn mice weakens antimicrobial host defenses. J Leukoc Biol 2025; 117:qiaf003. [PMID: 39824218 PMCID: PMC12022637 DOI: 10.1093/jleuko/qiaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/30/2024] [Accepted: 01/16/2025] [Indexed: 01/20/2025] Open
Abstract
Pediatric intensive care patients are particularly susceptible to severe bacterial infections because of ineffective neutrophil responses. The reasons why neutrophils of newborns are less responsive than those of adults are not clear. Because adenosine triphosphate and adenosine tightly regulate neutrophils, we studied whether the adenosine triphosphate and adenosine levels in the blood of newborn mice could impair the function of their neutrophils. We observed significant changes in plasma adenosine triphosphate and adenosine levels throughout the lifespan of mice. Adenosine levels in newborns were significantly higher than in older mice, while adenosine triphosphate levels were significantly lower. These changes were particularly striking in newborn and juvenile mice with adenosine triphosphate and adenosine levels of about 80 and 600 nM in newborns vs 130 and 190 nM in juveniles, respectively. The ratios of the adenosine triphosphate vs adenosine levels of newborns were (with 0.2) significantly lower than those of juveniles (1.4) and adults (0.5). These low adenosine triphosphate/adenosine ratios correlated with significantly weakened neutrophil activation responses following in vitro stimulation with a formyl peptide receptor agonist and a markedly higher morbidity and mortality rate of newborns following bacterial infection. We found that enhanced adenosine monophosphate hydrolysis via CD73, a lack of adenosine breakdown by adenosine deaminase, and reduced adenosine uptake by nucleoside transporters are responsible for the low adenosine triphosphate/adenosine ratios in blood of newborn mice. We conclude that the extracellular adenosine accumulation in newborn mice impairs inflammatory responses and reduces the ability of neutrophils to mount effective antimicrobial defenses against bacterial infections.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, University of California, San Diego Health, 9452 Medical Center Drive, La Jolla, CA 92037, United States
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Eleftheria-Angeliki Valsami
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Mark Elevado
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Ava Stevenson
- Department of Surgery, University of California, San Diego Health, 9452 Medical Center Drive, La Jolla, CA 92037, United States
| | - Reem Abutabikh
- Department of Surgery, University of California, San Diego Health, 9452 Medical Center Drive, La Jolla, CA 92037, United States
| | - Julian Curatolo
- Department of Surgery, University of California, San Diego Health, 9452 Medical Center Drive, La Jolla, CA 92037, United States
| | - Wolfgang G Junger
- Department of Surgery, University of California, San Diego Health, 9452 Medical Center Drive, La Jolla, CA 92037, United States
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| |
Collapse
|
123
|
Lu BT, Wang YJ, Wang L, Wu D, Lin YL, Xu JX, Zhang JN, Liu BW, Liu HY, Meng H, Gao Y, Wang HL, Kang K. Shenfu injection alleviates lipopolysaccharide-induced liver injury in septic mice. Sci Rep 2025; 15:14004. [PMID: 40263344 PMCID: PMC12015584 DOI: 10.1038/s41598-025-98740-3] [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: 07/30/2024] [Accepted: 04/14/2025] [Indexed: 04/24/2025] Open
Abstract
Shenfu injection (SFI) is a traditional Chinese medicine (TCM) for treating sepsis. The purpose of this study was to evaluate the protective effect of SFI on lipopolysaccharide (LPS)-induced liver injury in septic mice. The results showed that SFI intervention reduced liver/body weight and significantly improved the survival rate of septic mice. SFI could relieve the apoptosis of liver cells and ameliorate liver function in LPS-induced septic mice. SFI also diminished the serum and liver levels of the inflammatory factors IL-1β, IL-6, IL-18, IL-12, and TNF-α in a dose-dependent manner. SFI enhanced the mitochondrial membrane potential and alleviated the mitochondrial damage of liver in septic mice. Western blot revealed that the phosphorylation levels of IκB and NF-κB p65 increased significantly in the liver of LPS-induced septic mice. After SFI intervention, the phosphorylation levels of IκB and NF-κB p65 gradually recovered, especially at high concentration. SFI treatment reduced nuclear transduction, thus reducing transcriptional activity, which indicated that NF-κB p65 signal pathway might contribute to the anti-inflammatory and anti-apoptotic activities of SFI in the liver of LPS-induced septic mice. In addition, the metabolic profile of liver tissue in the model group was different from that in the control group, and SFI significantly regulated liver purine metabolism. These valuable findings suggested that SFI could improve mitochondrial function and mitigate inflammation and apoptosis, and thus alleviate LPS-induced liver injury in septic mice. SFI may be a promising drug to treat septic liver injury.
Collapse
Affiliation(s)
- Bai-Tao Lu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Ya-Jun Wang
- Department of Pediatrics, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, 150027, Heilongjiang Province, China
| | - Lei Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Di Wu
- Department of Critical Care Medicine, The Sixth Affiliated Hospital of Harbin Medical University, No.998 Aiying Road, Harbin, 150027, Heilongjiang Province, China
| | - Yi-Lu Lin
- Department of Critical Care Medicine, The Sixth Affiliated Hospital of Harbin Medical University, No.998 Aiying Road, Harbin, 150027, Heilongjiang Province, China
| | - Jia-Xi Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Jia-Ning Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Bo-Wen Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Hui-Ying Liu
- Department of Critical Care Medicine, The Sixth Affiliated Hospital of Harbin Medical University, No.998 Aiying Road, Harbin, 150027, Heilongjiang Province, China
| | - Huan Meng
- Department of Critical Care Medicine, The Sixth Affiliated Hospital of Harbin Medical University, No.998 Aiying Road, Harbin, 150027, Heilongjiang Province, China
| | - Yang Gao
- Department of Critical Care Medicine, The Sixth Affiliated Hospital of Harbin Medical University, No.998 Aiying Road, Harbin, 150027, Heilongjiang Province, China.
| | - Hong-Liang Wang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Road, Harbin, 150086, Heilongjiang Province, China.
| | - Kai Kang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
124
|
Wei N, Diekman CO. Dosing Time of Day Impacts the Safety of Antiarrhythmic Drugs in a Computational Model of Cardiac Electrophysiology. J Biol Rhythms 2025:7487304251326628. [PMID: 40269490 DOI: 10.1177/07487304251326628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Circadian clocks regulate many aspects of human physiology, including cardiovascular function and drug metabolism. Administering drugs at optimal times of the day may enhance effectiveness and reduce side effects. Certain cardiac antiarrhythmic drugs have been withdrawn from the market due to unexpected proarrhythmic effects such as fatal Torsade de Pointes (TdP) ventricular tachycardia. The Comprehensive in vitro Proarrhythmia Assay (CiPA) is a recent global initiative to create guidelines for the assessment of drug-induced arrhythmias that recommends a central role for computational modeling of ion channels and in silico evaluation of compounds for TdP risk. We simulated circadian regulation of cardiac excitability and explored how dosing time of day affects TdP risk for 11 drugs previously classified into risk categories by CiPA. The model predicts that a high-risk drug taken at the most optimal time of day may actually be safer than a low-risk drug taken at the least optimal time of day. Based on these proof-of-concept results, we advocate for the incorporation of circadian clock modeling into the CiPA paradigm for assessing drug-induced TdP risk. Since cardiotoxicity is the leading cause of drug discontinuation, modeling cardiac-related chronopharmacology has significant potential to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Ning Wei
- Department of Mathematics, Purdue University, West Lafayette, Indiana
| | - Casey O Diekman
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, New Jersey
| |
Collapse
|
125
|
Ruan W, Li T, Bang IH, Lee J, Deng W, Ma X, Luo C, Du F, Yoo SH, Kim B, Li J, Yuan X, Figarella K, An YA, Wang YY, Liang Y, DeBerge M, Zhang D, Zhou Z, Wang Y, Gorham JM, Seidman JG, Seidman CE, Aranki SF, Nair R, Li L, Narula J, Zhao Z, Gorfe AA, Muehlschlegel JD, Tsai KL, Eltzschig HK. BMAL1-HIF2A heterodimer modulates circadian variations of myocardial injury. Nature 2025:10.1038/s41586-025-08898-z. [PMID: 40269168 DOI: 10.1038/s41586-025-08898-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/14/2025] [Indexed: 04/25/2025]
Abstract
Acute myocardial infarction is a leading cause of morbidity and mortality worldwide1. Clinical studies have shown that the severity of cardiac injury after myocardial infarction exhibits a circadian pattern, with larger infarcts and poorer outcomes in patients experiencing morning-onset events2-7. However, the molecular mechanisms underlying these diurnal variations remain unclear. Here we show that the core circadian transcription factor BMAL17-11 regulates circadian-dependent myocardial injury by forming a transcriptionally active heterodimer with a non-canonical partner-hypoxia-inducible factor 2 alpha (HIF2A)12-16-in a diurnal manner. To substantiate this finding, we determined the cryo-EM structure of the BMAL1-HIF2A-DNA complex, revealing structural rearrangements within BMAL1 that enable cross-talk between circadian rhythms and hypoxia signalling. BMAL1 modulates the circadian hypoxic response by enhancing the transcriptional activity of HIF2A and stabilizing the HIF2A protein. We further identified amphiregulin (AREG)16,17 as a rhythmic target of the BMAL1-HIF2A complex, critical for regulating daytime variations of myocardial injury. Pharmacologically targeting the BMAL1-HIF2A-AREG pathway provides cardioprotection, with maximum efficacy when aligned with the pathway's circadian phase. These findings identify a mechanism governing circadian variations of myocardial injury and highlight the therapeutic potential of clock-based pharmacological interventions for treating ischaemic heart disease.
Collapse
Affiliation(s)
- Wei Ruan
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, China.
| | - Tao Li
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - In Hyuk Bang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jaewoong Lee
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Wankun Deng
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xinxin Ma
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Cong Luo
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Fang Du
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Boyun Kim
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Major in Aquaculture and Applied Life Sciences, College of Fisheries Science, Pukyong National University, Busan, Republic of Korea
| | - Jiwen Li
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Department of Cardiac Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyi Yuan
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Katherine Figarella
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Yin-Ying Wang
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yafen Liang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Center for Outcomes Research, UTHealth Houston, Houston, TX, USA
| | - Matthew DeBerge
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Dongze Zhang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Zhen Zhou
- Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Yanyu Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Joshua M Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | | | - Sary F Aranki
- Department of Surgery, Division of Cardiac Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ragini Nair
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Lei Li
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jagat Narula
- Division of Cardiology, Department of Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Memorial Hermann Hospital, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alemayehu A Gorfe
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jochen D Muehlschlegel
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kuang-Lei Tsai
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.
- MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA.
| | - Holger K Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.
- Center for Outcomes Research, UTHealth Houston, Houston, TX, USA.
| |
Collapse
|
126
|
Khan MG, Hussain SHA, Alkhayl FFA, Ahsan M, Ridha-Salman H. Cannabinoids in neuropathic pain treatment: pharmacological insights and clinical outcomes from recent trials. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04134-7. [PMID: 40261351 DOI: 10.1007/s00210-025-04134-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/01/2025] [Indexed: 04/24/2025]
Abstract
Neuropathic pain, a complex and often devastating condition, poses significant challenges for its effective management. Despite promising research on various cannabis formulations and delivery methods for neuropathic pain, significant gaps remain in our knowledge. While inhaled cannabis shows analgesic effects and alternative delivery methods may improve bioavailability, oral formulations have yielded mixed results, often limited by small sample sizes and placebo effects. Therefore, further research is essential to optimize cannabis formulations, identify responder profiles to tailor treatments effectively, and, most critically, confirm the long-term safety and efficacy of cannabis-based therapies in managing NP. This review article aims to provide a comprehensive analysis of the therapeutic potential of cannabis-based medicines, with a particular focus on cannabinoids. This review, though not systematic, examines 11 clinical studies, specifically Randomised Clinical Trials) published from 2014 to 2024, highlighting the efficacy of numerous cannabis formulations, in alleviating neuropathic pain. Key findings show that cannabinoids can reduce pain perception, improve patient quality of life, and mitigate other symptoms associated with neuropathic pain. The synergistic effects of tetrahydrocannabinol and cannabidiol are discussed, emphasizing their ability to enhance analgesic effects, while potentially reducing the psychoactive side effects of tetrahydrocannabinol. This review emphasizes the importance of the personalized approach to improve therapeutic outcomes. Limitations of the existing research focusing on cannabis for neuropathic pain are limited by heterogeneity, lack of standardization, small sample sizes, and reliance on subjective outcomes, impacting the reliability and generalizability of findings. However, this exhaustive review aims to inform clinicians and researchers about the evolving role of cannabis in contemporary pain management strategies, illustrating the diverse pharmacological profiles of cannabinoids and their potential as adjunct therapies for neuropathic pain management.
Collapse
Affiliation(s)
- Mohammad Gayoor Khan
- Department of Pharmacology, Daksh Institute of Pharmaceutical Science, Chhatarpur Madhya Pradesh, General Secretary Society of Clinical Research and Medical Professionals, Hyderabad, 500059, India
| | - Sajid Hussain Altaf Hussain
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, 11952, Majmaah, Saudi Arabia.
| | - Faris F Aba Alkhayl
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Marya Ahsan
- Department of Pharmacology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Hayder Ridha-Salman
- College of Pharmacy, Department of Pharmacology, Al-Mustaqbal University, 51001, Babylon, Iraq
| |
Collapse
|
127
|
Szabo C. Alcohol and the aging cardiovascular system: a dangerous synergy uncovered. GeroScience 2025:10.1007/s11357-025-01644-3. [PMID: 40261452 DOI: 10.1007/s11357-025-01644-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/24/2025] Open
Affiliation(s)
- Csaba Szabo
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
128
|
Zhong C, Deng K, Lang X, Shan D, Xie Y, Pan W, Yu J. Therapeutic potential of natural flavonoids in atherosclerosis through endothelium-protective mechanisms: An update. Pharmacol Ther 2025; 271:108864. [PMID: 40274196 DOI: 10.1016/j.pharmthera.2025.108864] [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: 12/10/2024] [Revised: 02/27/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
Atherosclerosis and its associated cardiovascular complications remain significant global public health challenges, underscoring the urgent need for effective therapeutic strategies. Endothelial cells are critical for maintaining vascular health and homeostasis, and their dysfunction is a key contributor to the initiation and progression of atherosclerosis. Targeting endothelial dysfunction has, therefore, emerged as a promising approach for the prevention and management of atherosclerosis. Among natural products, flavonoids, a diverse class of plant-derived phenolic compounds, have garnered significant attention for their anti-atherosclerotic properties. A growing body of evidence demonstrates that flavonoids can mitigate endothelial dysfunction, highlighting their potential as endothelial dysfunction-targeted therapeutics for atherosclerosis. In this review, we summarize current knowledge on the roles of natural flavonoids in modulating various aspects of endothelial dysfunction and their therapeutic effects on atherosclerosis, focusing on the underlying molecular mechanisms. We also discuss the challenges and future prospects of translating natural flavonoids into clinical applications for cardiovascular medicine. This review aims to provide critical insights to advance the development of novel endothelium-protective pharmacotherapies for atherosclerosis.
Collapse
Affiliation(s)
- Chao Zhong
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Keke Deng
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xiaoya Lang
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Dan Shan
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Yanfei Xie
- Center for Translational Medicine, Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wen Pan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| | - Jun Yu
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
129
|
Barnes DA, Janssen MJ, Yang H, Redegeld FA, Masereeuw R. An adverse outcome pathway for DNA adduct formation leading to kidney failure. Toxicology 2025; 515:154162. [PMID: 40268266 DOI: 10.1016/j.tox.2025.154162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/11/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
An Adverse Outcome Pathway (AOP) is a conceptual framework in toxicology and risk assessment that outlines the series of events from a chemical's molecular interaction to the resulting adverse health effect. This framework offers a structured approach to organizing biological knowledge, making it especially useful for understanding the mechanisms through which chemicals cause harm. Following a comprehensive analysis of the literature, an AOP was elucidated for key events linking DNA adduct formation, caused by compounds such as platinum anticancer drugs, to tubular necrosis, resulting in kidney failure. Currently, cisplatin, carboplatin and oxaliplatin are the three most utilised Pt-based drugs used globally for the treatment of cancer. The hydrolysis of platinum anticancer agents post-cellular uptake yields electrophilic intermediates that covalently bind to nucleophilic sites on DNA to form adducts that represent the molecular initiating event. When DNA repair mechanisms become unbalanced, the nephrotoxic response following the formation of DNA adducts leads to DNA damage and mitochondrial dysfunction. These events promote the generation and release of reaction oxygen species (ROS) to induce oxidative stress, causing cell death and inflammation. Upon detachment from the basement membrane, these compromised cells are subsequently deposited in the tubular lumen. Tubular obstruction and inflammatory responses to proximal tubule insult can lead to secondary toxicity and tubular necrosis, further exacerbating kidney injury and precipitating a progressive decline of renal function, finally resulting in kidney failure.
Collapse
Affiliation(s)
- D A Barnes
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands
| | - M J Janssen
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands
| | | | - F A Redegeld
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands
| | - R Masereeuw
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands.
| |
Collapse
|
130
|
Slominski RM, Raman C, Jetten AM, Slominski AT. Neuro-immuno-endocrinology of the skin: how environment regulates body homeostasis. Nat Rev Endocrinol 2025:10.1038/s41574-025-01107-x. [PMID: 40263492 DOI: 10.1038/s41574-025-01107-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 04/24/2025]
Abstract
The skin, including the hypodermis, is the largest organ of the body. The epidermis, the uppermost layer, is in direct contact with the environment and is exposed to environmental stressors, including solar radiation and biological, chemical and physical factors. These environmental factors trigger local responses within the skin that modulate homeostasis on both the cutaneous and systemic levels. Using mediators in common with brain pathways, immune and neuroendocrine systems within the skin regulate these responses to activate various signal transduction pathways and influence the systemic endocrine and immune systems in a context-dependent manner. This skin neuro-immuno-endocrine system is compartmentalized through the formation of epidermal, dermal, hypodermal and adnexal regulatory units. These units can act separately or in concert to preserve skin integrity, allow for adaptation to a changing environment and prevent the development of pathological processes. Through activation of peripheral nerve endings, the release of neurotransmitters, hormones, neuropeptides, and cytokines and/or chemokines into the circulation, or by priming circulating and resident immune cells, this system affects central coordinating centres and global homeostasis, thus adjusting the body's homeostasis and allostasis to optimally respond to the changing environment.
Collapse
Affiliation(s)
- Radomir M Slominski
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chander Raman
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anton M Jetten
- Cell Biology Section, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Andrzej T Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL, USA.
- Veteran Administration Medical Center, Birmingham, AL, USA.
| |
Collapse
|
131
|
Lee JJW, Hamour AF, Wihlidal JGJ, Lee JM, Monteiro E, Poduch E, Kotra L, Vescan AD. Cannabinoids and the Endocannabinoid System in the Treatment of Chronic Rhinosinusitis. Laryngoscope 2025. [PMID: 40260746 DOI: 10.1002/lary.32191] [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: 11/26/2024] [Revised: 02/25/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVE Recently, the endocannabinoid system (ECS) has emerged as a therapeutic target for various inflammatory diseases, including those of the respiratory tract. The objective of this scoping review is to explore the role of the ECS in the pathophysiology of CRS. Moreover, we sought to identify, appraise, and summarize the available evidence for cannabinoids as a potential treatment for CRS. DATA SOURCES Six databases and four clinical trial registries were searched from inception to February 2025. REVIEW METHODS All identified studies investigating the role of the ECS in sinonasal inflammatory disease were included for review. RESULTS A total of 1534 studies were identified in the initial search. Following screening and full-text analysis by three authors, five studies were included in the final scoping review. Four of the studies were preclinical and in vitro in nature, examining the effects of ECS modulation through CB1 and/or CB2 receptors. The findings of each study support a common conclusion that the ECS is implicated in regulating cellular inflammatory pathways potentially involved in sinonasal disease. The final study investigated the effect of marijuana smoking on subjective and objective measures of CRS severity. There were no clinical studies identified investigating the use of cannabinoids for the treatment of sinonasal inflammatory conditions. CONCLUSION Current literature examining the role of ECS in sinonasal inflammatory disease is highly limited, though it indicates ECS may play a role in the complex pathophysiology of sinonasal inflammatory disease. Further work is warranted to assess ECS as a potential therapeutic target for CRS.
Collapse
Affiliation(s)
- John J W Lee
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Amr F Hamour
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Jacob G J Wihlidal
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - John M Lee
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Eric Monteiro
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Ewa Poduch
- Center for Molecular Design and Preformulations, Toronto General Hospital Research Institute, University of Toronto, Toronto, Canada
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Lakshmi Kotra
- Center for Molecular Design and Preformulations, Toronto General Hospital Research Institute, University of Toronto, Toronto, Canada
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Allan D Vescan
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
132
|
Ghosh R, Mazumdar R, Samanta B, Saha S, Mondal B. Reaction of a non-heme iron-nitrosyl with dioxygen: decomposition of the ligand through NOD-like activity. Dalton Trans 2025. [PMID: 40260957 DOI: 10.1039/d5dt00009b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
A high-spin iron(II) nitrosyl, [(TPz)Fe(NO)](ClO4)2, 2 (TPz = Tris(3,5-dimethylpyrazol-1-ylmethyl)amine) with an {Fe(NO)}7 configuration was synthesized and characterized structurally. The dioxygen reactivity of complex 2 in acetonitrile solution results in the oxidation of the ligand. Chemical evidence suggests the involvement of a peroxynitrite intermediate in this reaction. A trapping experiment shows the formation of NO2 during the reaction which supports the proposition of the involvement of the peroxynitrite intermediate. This study gives an insight into an alternate possibility from the dioxygen reactivity of metal-nitrosyl leading to nitric oxide dioxygenase (NOD) activity.
Collapse
Affiliation(s)
- Riya Ghosh
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam - 781039, India.
| | - Rakesh Mazumdar
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam - 781039, India.
| | - Bapan Samanta
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam - 781039, India.
| | - Shankhadeep Saha
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam - 781039, India.
| | - Biplab Mondal
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam - 781039, India.
| |
Collapse
|
133
|
Tschann MM, Vachharajani V, Redmond EM, Hoisington A, Cohen SE, New-Aaron M, Llorente C, Paloczi J, Keating CR, Rungratanawanich W, Burnham EL, Callaci JJ, Raju P, Zhong W, Mandal A, Zimmerly JR, Nuncio ASP, Mandrekar P, McCullough RL, McMahan RH, Wyatt TA, Yeligar SM, Kovacs EJ, Choudhry MA. New developments on the effects of alcohol use on immunity, inflammation and organ function: A summary of the 2024 Alcohol and Immunology Research Interest Group (AIRIG) meeting. Alcohol 2025; 126:1-10. [PMID: 40267994 DOI: 10.1016/j.alcohol.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
The 29th annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held on November 22nd, 2024, at Loyola University Chicago, Health Science Campus, Maywood, Illinois. The meeting was divided into three plenary sessions and a poster session. The overall focus of this year's meeting was on alcohol and host immunity, alcohol and organ dysfunction, and alcohol, inflammation, and tissue injury. The presentations in each session shared the latest developments on the impact of alcohol in a wide variety of fields including trauma, emergency care and hospitalization, cardiovascular health, neurodegenerative disease, gut microbiome, and hepatology.
Collapse
Affiliation(s)
- Madison M Tschann
- Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA; Alcohol Research Program, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | | | - Eileen M Redmond
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Andrew Hoisington
- Department of Preventative Medicine & Rehabilitation, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Sarah E Cohen
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Moses New-Aaron
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, and Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Janos Paloczi
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Claudia R Keating
- Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA; Alcohol Research Program, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA; Department of Cancer Biology, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL, USA
| | - Wiramon Rungratanawanich
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Ellen L Burnham
- Alcohol Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - John J Callaci
- Alcohol Research Program, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA; Department of Orthopaedic Surgery and Rehabilitation, Loyola University Chicago, Health Sciences Campus, Maywood, IL, USA
| | - Preeti Raju
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Weizhe Zhong
- Division of Digestive Disease, Internal Medicine, Yale University, New Haven, CT, USA; Yale Liver Center, New Haven, CT, USA
| | - Abhishek Mandal
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Justine R Zimmerly
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Adriana S P Nuncio
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Pranoti Mandrekar
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Rebecca L McCullough
- Alcohol Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rachel H McMahan
- Alcohol Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Division of GI, Trauma and Endocrine Surgery, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Todd A Wyatt
- Pulmonary Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Department of Environmental, Agricultural and Occupational Health, College of Public Health, Omaha, NE, USA
| | - Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, and Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
| | - Elizabeth J Kovacs
- Alcohol Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Division of GI, Trauma and Endocrine Surgery, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Veterans Health Administration, Eastern Colorado Health Care System, Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, USA
| | - Mashkoor A Choudhry
- Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA; Alcohol Research Program, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA.
| |
Collapse
|
134
|
Marcos-Ríos D, Rochano-Ortiz A, San Sebastián-Jaraba I, Fernández-Gómez MJ, Méndez-Barbero N, Oller J. Mitochondrial Dysfunction: A New Hallmark in Hereditable Thoracic Aortic Aneurysm Development. Cells 2025; 14:618. [PMID: 40277943 PMCID: PMC12026022 DOI: 10.3390/cells14080618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
Thoracic aortic aneurysms (TAAs) pose a significant health burden due to their asymptomatic progression, often culminating in life-threatening aortic rupture, and due to the lack of effective pharmacological treatments. Risk factors include elevated hemodynamic stress on the ascending aorta, frequently associated with hypertension and hereditary genetic mutations. Among the hereditary causes, Marfan syndrome is the most prevalent, characterized as a connective tissue disorder driven by FBN1 mutations that lead to life-threatening thoracic aortic ruptures. Similarly, mutations affecting the TGF-β pathway underlie Loeys-Dietz syndrome, while mutations in genes encoding extracellular or contractile apparatus proteins, such as ACTA2, are linked to non-syndromic familial TAA. Despite differences in genetic origin, these hereditary conditions share central pathophysiological features, including aortic medial degeneration, smooth muscle cell dysfunction, and extracellular remodeling, which collectively weaken the aortic wall. Recent evidence highlights mitochondrial dysfunction as a crucial contributor to aneurysm formation in Marfan syndrome. Disruption of the extracellular matrix-mitochondrial homeostasis axis exacerbates aortic wall remodeling, further promoting aneurysm development. Beyond its structural role in maintaining vascular integrity, the ECM plays a pivotal role in supporting mitochondrial function. This intricate relationship between extracellular matrix integrity and mitochondrial homeostasis reveals a novel dimension of TAA pathophysiology, extending beyond established paradigms of extracellular matrix remodeling and smooth muscle cell dysfunction. This review summarizes mitochondrial dysfunction as a potential unifying mechanism in hereditary TAA and explores how understanding mitochondrial dysfunction, in conjunction with established mechanisms of TAA pathogenesis, opens new avenues for developing targeted treatments to address these life-threatening conditions. Mitochondrial boosters could represent a new clinical opportunity for patients with hereditary TAA.
Collapse
Affiliation(s)
- Daniel Marcos-Ríos
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
| | - Antonio Rochano-Ortiz
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
| | - Irene San Sebastián-Jaraba
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - María José Fernández-Gómez
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
| | - Nerea Méndez-Barbero
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Facultad de Medicina, Universidad Alfonso X el Sabio (UAX), Villanueva de la Cañada, 28691 Madrid, Spain
| | - Jorge Oller
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Facultad de Medicina, Universidad Alfonso X el Sabio (UAX), Villanueva de la Cañada, 28691 Madrid, Spain
| |
Collapse
|
135
|
Unlu Y, Stinson EJ, Krakoff J, Piaggi P. Protein oxidation in non-exercising healthy adults under varying dietary conditions: Physiological determinants, effects on fuel partitioning, and implications for body weight regulation. Metabolism 2025; 169:156270. [PMID: 40268049 DOI: 10.1016/j.metabol.2025.156270] [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: 01/09/2025] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Protein oxidation (PROTOX) typically accounts for the smallest fraction of daily energy expenditure (24hEE) in humans compared to carbohydrate and lipid oxidation. However, inter-individual differences in PROTOX may explain differences in fuel partitioning and body weight change. We aimed to elucidate the physiological determinants of PROTOX under controlled 24-h dietary conditions, including eucaloric feeding, fasting, and overfeeding diets with variable protein content. METHODS Eighty-six weight-stable healthy volunteers with normal glucose regulation (67 M/19F; age: 37 ± 10 years; BMI: 26.7 ± 4.5 kg/m2, body fat by DXA: 29.0 ± 9.8 %) underwent 24hEE measurements by whole-room calorimetry during energy balance (20 % protein, 50 % carbohydrate), different overfeeding diets (200 % of the daily eucaloric requirement), including three normal-protein (20 %) diets (balanced: 50 % carbohydrate; high-carbohydrate: 75 % carbohydrate; high-fat: 60 % fat), low-protein (3 %) and high-protein (30 %), and 24-h fasting in a randomized crossover design. Urine samples were collected during each 24-h dietary intervention for quantification of PROTOX and catecholamine excretion rates by nitrogen excretion and high-performance liquid chromatography, respectively. RESULTS PROTOX during energy balance (mean ± SD: 372 ± 78 kcal/day) was positively associated with protein intake (r = 0.39, p < 0.001), fat free mass (r = 0.35, p < 0.001), but not with fat mass (p = 0.24). Higher PROTOX was associated with higher 24-h urinary norepinephrine (partial r = 0.27, p = 0.01), but not epinephrine (p = 0.48), excretion rates. During normal-protein diets, higher PROTOX was associated with lower lipid oxidation, but showed no association with carbohydrate oxidation. Inter-individual variability in PROTOX did not predict changes in weight or body composition over two years. CONCLUSION Dietary protein content, lean body mass, and sympathetic nervous system activity are key determinants of PROTOX. Although PROTOX did not predict free-living weight gain, increased PROTOX is associated with decreased lipid oxidation, underscoring its role in fuel partitioning and whole-body energy and substrate balance.
Collapse
Affiliation(s)
- Yigit Unlu
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ, United States of America
| | - Emma J Stinson
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ, United States of America
| | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ, United States of America
| | - Paolo Piaggi
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ, United States of America; Department of Information Engineering, University of Pisa, Pisa, Italy.
| |
Collapse
|
136
|
Sahin KE, Karatas M, Barutcu S, Inanc IH. Evaluation of the Electrocardiographic Tp-e, Tp-e/QT, and Tp-e/QTc Parameters in Patients with Non-Alcoholic Liver Disease. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:766. [PMID: 40283057 PMCID: PMC12028712 DOI: 10.3390/medicina61040766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/04/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Non-alcoholic fatty liver disease (NAFLD) is a common chronic liver disease associated with significant morbidity, including cardiovascular complications. This study investigates the relationship between NAFLD and electrocardiographic parameters indicative of ventricular arrhythmia risk. Materials and Methods: We conducted a cross-sectional study enrolling 136 patients with NAFLD and 136 healthy controls. Electrocardiographic parameters-Tp-e interval, QT and corrected QT (QTc) intervals, and Tp-e/QTc ratio-were measured and compared between groups. Results: Patients with NAFLD exhibited significantly higher Tp-e, QTc, Tp-e/QT ratio, and Tp-e/QTc ratio (p < 0.001, for all) than controls. Subgroup analysis showed progressive increases in Tp-e and Tp-e/QT ratio correlating with NAFLD severity (p < 0.001 and p = 0.001, respectively, for grade 1 vs. grade 2; p < 0.001 and p = 0.001, respectively, for grade 1 vs. grade 3). ROC analysis indicated that the Tp-e interval was a strong predictor for identifying grade 2 or more NAFLD (AUC 0.887, p < 0.001). Conclusions: Our findings highlight the association of NAFLD with prolonged electrocardiographic intervals that may predispose patients to ventricular arrhythmias. These parameters can serve as valuable markers for cardiac risk stratification in patients with NAFLD, suggesting the need for vigilant cardiac follow-up in this population.
Collapse
Affiliation(s)
- Kader Eliz Sahin
- Department of Cardiology, Kocaeli City Hospital, Kocaeli 41060, Turkey
| | - Mesut Karatas
- Department of Cardiology, Kosuyolu High Specialization Education and Research Hospital, Istanbul 34865, Turkey;
| | - Sezgin Barutcu
- Department of Gastroenterology, Gaziantep University Faculty of Medicine, Gaziantep 27310, Turkey;
| | - Ibrahim Halil Inanc
- Department of Cardiology, Kirikkale Yuksek Ihtisas Hospital, Kirikkale 27310, Turkey;
- Department of Cardiology, Phoenixville Hospital—Tower Health, Phoenixville, PA 19460, USA
| |
Collapse
|
137
|
Simankowicz P, Stępniewska J. The Role of Endocannabinoids in Physiological Processes and Disease Pathology: A Comprehensive Review. J Clin Med 2025; 14:2851. [PMID: 40283681 PMCID: PMC12027566 DOI: 10.3390/jcm14082851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/18/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025] Open
Abstract
The endocannabinoid system is a complex communication system involved in maintaining homeostasis in various physiological processes, including metabolism, immune response, pain modulation, and neuroprotection. Endocannabinoids, mainly anandamide and 2-arachidonoylglycerol, are natural ligands of the cannabinoid receptors CB1 and CB2, which are widely distributed throughout the central nervous system and peripheral tissues. Their biosynthesis, degradation, and interaction with other signaling pathways play crucial roles in both health and disease. This article provides a comprehensive overview of the physiological and pathological roles of endocannabinoids, discusses their potential as therapeutic targets, and highlights recent advances in endocannabinoid-based treatments.
Collapse
Affiliation(s)
| | - Joanna Stępniewska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland
| |
Collapse
|
138
|
Liang P, Chen JJ, Yang X, Long R, Li Y, Wang ZL, Yang PL, Liang YD. Association and functional study of ATP6V1D and GPHN gene polymorphisms with depression in Chinese population. World J Psychiatry 2025; 15:102182. [PMID: 40309610 PMCID: PMC12038669 DOI: 10.5498/wjp.v15.i4.102182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Depression is a disease with a significant global social burden. Single nucleotide polymorphisms (SNPs) are correlated with the development of depression. This study investigates the relationship between polymorphisms in the GPHN and ATP6V1D gene promoter regions and susceptibility to depression in the Chinese population. AIM To provide new insights into identifying SNPs for predicting depression in the Chinese population. METHODS We conducted a case-control study involving 555 individuals with depression and 509 healthy controls. GPHN rs8020095 and ATP6V1D rs3759755, rs10144417, rs2031564, and rs8016024 in the promoter region were genotyped using next-generation sequencing. Dual luciferase reporter genes were employed to assess the transcriptional activity of promoter regions for each SNP genotype, with transcription factors binding to each site predicted using the JASPAR database. RESULTS Compared to healthy controls, the ATP6V1D promoter rs10144417 AG genotype (P = 0.015), rs3759755 AC/CC genotype (P = 0.036), and GPHN gene rs8020095 GA and AA genotypes (GA: P = 0.028, GG: P = 0.025) were significantly associated with a lower prevalence of depression. Linked disequilibria were present in five SNPs, with the AGATA haplotype frequency in patients significantly lower than in healthy subjects (P = 0.023). Luciferase activity of the rs3759755-A recombinant plasmid was significantly higher than that of the rs3759755-C recombinant plasmid (P = 0.026), and the rs8020095-A recombinant plasmid activity was significantly higher than that of the rs8020095-G recombinant plasmid (P = 0.001). Transcription factors orthodenticle homeobox 2, orthodenticle homeobox 1, forkhead box L1, NK homeobox 3-1, and nuclear factor, interleukin 3 regulated demonstrated binding affinity with rs3759755A > C and rs8020095A > G. CONCLUSION This study demonstrates that SNPs (rs3759755 and rs10144417) in the promoter region of the ATP6V1D and SNP (rs8020095) of GPHN are indeed associated with susceptibility to depression.
Collapse
Affiliation(s)
- Peng Liang
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Jing-Jie Chen
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Xue Yang
- Department of Geriatric Psychiatry, The First Psychiatric Hospital of Harbin, Harbin 150001, Heilongjiang Province, China
| | - Rui Long
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Yue Li
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Zi-Ling Wang
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Ping-Liang Yang
- Department of Anesthesiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Yun-Dan Liang
- Department of Basic Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| |
Collapse
|
139
|
Fu L, Yokus B, Gao B, Pacher P. An Update on IL-22 Therapies in Alcohol-Associated Liver Disease and Beyond. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00117-8. [PMID: 40254130 DOI: 10.1016/j.ajpath.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/22/2025]
Abstract
Excessive alcohol consumption drives the development of alcohol-associated liver disease (ALD), including steatohepatitis, cirrhosis, and hepatocellular carcinoma, and its associated complications, such as hepatorenal syndrome. Hepatocyte death, inflammation, and impaired liver regeneration are key processes implicated in the pathogenesis and progression of ALD. Despite extensive research, therapeutic options for ALD remain limited. IL-22 has emerged as a promising therapeutic target because of its hepatoprotective properties mediated through the activation of the STAT3 signaling pathway. IL-22 enhances hepatocyte survival by mitigating apoptosis, oxidative stress, and inflammation while simultaneously promoting liver regeneration through the proliferation of hepatocytes and hepatic progenitor cells and the up-regulation of growth factors. Additionally, IL-22 exerts protective effects on epithelial cells in various organs affected by ALD and its associated complications. Studies from preclinical models and early-phase clinical trials of IL-22 agonists, such as F-652 and UTTR1147A, have shown favorable safety profiles, good tolerability, and encouraging efficacy in reducing liver injury and promoting regeneration. However, the heterogeneity and multifactorial nature of ALD present ongoing challenges. Further research is needed to optimize IL-22-based therapies and clarify their roles within a comprehensive approach to ALD management. This review summarizes the current understanding of IL-22 biology and its role in ALD pathophysiology and ALD-associated complications along with therapeutic application of IL-22, potential benefits, and limitations.
Collapse
Affiliation(s)
- Lihong Fu
- Laboratory of Cardiovascular Physiology and Tissue Injury, NIH/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Burhan Yokus
- Laboratory of Cardiovascular Physiology and Tissue Injury, NIH/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Bin Gao
- Laboratory of Liver Diseases, NIH/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland.
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, NIH/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland.
| |
Collapse
|
140
|
Arnold W, Jain S, Sinha V, Das A. The Hunt for the Putative Epoxyeicosatrienoic Acid Receptor. ACS Chem Biol 2025; 20:762-777. [PMID: 40127470 PMCID: PMC12012780 DOI: 10.1021/acschembio.5c00047] [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: 01/15/2025] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/26/2025]
Abstract
Epoxyeicosatrienoic acids, or EETs, are signaling molecules formed by the metabolism of arachidonic acid by cytochrome P450 enzymes. They are well-known for their anti-inflammatory effects, their ability to lower blood pressure, and benefits to cardiovascular outcomes. Despite the wealth of data demonstrating their physiological benefits, the putative high-affinity receptor that mediates these effects is yet to be identified. The recent report that the sphingosine-1-phosphate receptor 1 (S1PR1) is a high-affinity receptor for a related epoxy lipid prompted us to ask, "Why has the putative EET receptor not been discovered yet? What information about the discoveries of lipid epoxide receptors can help us identify the putative EET receptor?" In this review, we summarize the evidence supporting that the putative EET receptor exists. We then review the data showing EETs binding to other, low-affinity receptors and the discovery of receptors for similar lipid metabolites that can serve as a model for identifying the putative EET receptor. We hope this review will revitalize the search for this important receptor, which can facilitate the development of anti-inflammatory and cardiovascular therapeutics.
Collapse
Affiliation(s)
- William
R. Arnold
- Stanford
Cryo-EM Center, Stanford University School
of Medicine, Palo Alto, California 94305, United States
| | - Sona Jain
- Departamento
de Morfologia, Universidade Federal de Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Vidya Sinha
- The
Center for Advanced Studies in Science, Math and Technology at Wheeler
High School, Marietta, Georgia 30068, United States
| | - Aditi Das
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology (GaTech), Atlanta, Georgia 30332, United States
| |
Collapse
|
141
|
Mangutov E, Awad-Igbaria Y, Siegersma K, Gastambide F, Asuni AA, Pradhan AAA. Enhancement of the endocannabinoid system through monoacylglycerol lipase inhibition relieves migraine-associated pain in mice. J Headache Pain 2025; 26:84. [PMID: 40251497 PMCID: PMC12007319 DOI: 10.1186/s10194-025-02029-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Migraine affects over 1 billion people worldwide and is a leading cause of disability. Targeting the cannabinoid system offers a promising approach for pain and migraine relief. This study evaluated a novel monoacylglycerol lipase (MAGL) inhibitor to prolong endocannabinoid action in acute and chronic mouse models of migraine. It also examined MAGL and cannabinoid receptor 1 (CB1) mRNA expression in key head pain-processing regions. METHODS C57BL6/J male and female mice received the human migraine trigger nitroglycerin (NTG) acutely or every other day for 9 days. Allodynia was assessed by von Frey hair stimulation of the periorbital area. A single dose of MAGL inhibitor (ABD-1970) was tested in acute and chronic NTG models. Additionally, ABD-1970 was given daily for 5 days to assess tolerance. In situ hybridization measured transcript expression of MAGL, CB1, and neuronal marker Rbfox3 in trigeminal ganglia (TG) and trigeminal nucleus caudalis (TNC). RESULTS A single injection of ABD-1970 blocked cephalic allodynia induced by acute NTG. ABD-1970 also blocked chronic allodynia established by chronic intermittent NTG. Repeated administration did not induce tolerance, and ABD-1970 continued to block NTG-induced allodynia after 5 days of administration. There was high expression of MAGL and CB1 in the TG and TNC, present in Rbfox3 positive and negative cells. CONCLUSION MAGL inhibitor effectively blocked acute and chronic migraine-associated pain, likely through prolonged endocannabinoid action. This effect may be mediated through action at peripheral or central sites considering the high MAGL and CB1 expression in the TG and TNC, respectively. The endocannabinoid system appears to modulate migraine mechanisms, and MAGL may be a promising target for this disorder.
Collapse
Affiliation(s)
- Elizaveta Mangutov
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Yaseen Awad-Igbaria
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kendra Siegersma
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | | | | | - Amynah A A Pradhan
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
142
|
Tang LF, Tang FL, Zhou H, Li ZK, Pi CQ, He Y, Li M. Bacillus Coagulans BC99 Protects Ionizing Radiation-Induced Intestinal Injury and Modulates Gut Microbiota and Metabolites in Mice. Mol Nutr Food Res 2025:e70057. [PMID: 40243794 DOI: 10.1002/mnfr.70057] [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: 12/12/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025]
Abstract
The gastrointestinal tract is highly sensitive to ionizing radiation (IR), which causes radiation-induced intestinal injury (RIII). There are no effective drugs available for RIII in routine clinical treatment, which is a major limiting factor during the process of radiotherapy for pelvic abdominal malignancies. In this study, we aimed to elucidate the potential of probiotic Bacillus coagulans BC99 (B.coagulans BC99) in preventing RIII. C57BL/6J mice were gavage-administered with B.coagulans BC99 for 30 days and then exposed to a single dose of 12 Gy x-ray whole abdominal irradiation (WAI). B.coagulans BC99 treatment could mitigate RIII by preventing weight loss, maintaining the integrity of intestinal structure and barrier, improving inflammatory symptoms, modulating oxidative stress, and regulating the composition of gut microbiota, thereby reestablishing intestinal homeostasis. In addition, the potential radioprotective mechanism of B.coagulans BC99 was closely related to the gut microbiota-derived metabolites. This study offers a novel perspective for advancing probiotic-based treatments for RIII and enhancing strategies for the prevention of RIII.
Collapse
Affiliation(s)
- Lin-Feng Tang
- State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng-Ling Tang
- Department of Oncology and Hematology, The Zhongxian People's Hospital, Chongqing, China
| | - Hao Zhou
- State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Ze-Kun Li
- State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Chao-Qun Pi
- MOE Engineering Center of Hematological Disease, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yang He
- MOE Engineering Center of Hematological Disease, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Ming Li
- State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| |
Collapse
|
143
|
Tortolani D, Decandia D, Giacovazzo G, Scipioni L, Panuccio A, Ciaramellano F, Eugelio F, Fanti F, Latagliata EC, La Barbera L, Cutuli D, Compagnone D, D’Amelio M, Coccurello R, Oddi S, Petrosini L, Maccarrone M. Chronic palmitoylethanolamide administration via slow-release subcutaneous pellets promotes neuroprotection and mitigates neuroinflammation in the Tg2576 mouse model of Alzheimer's disease. Front Cell Neurosci 2025; 19:1571428. [PMID: 40313591 PMCID: PMC12043567 DOI: 10.3389/fncel.2025.1571428] [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: 02/05/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive and non-cognitive decline associated with neuropathological hallmarks, including neuroinflammation. Palmitoylethanolamide (PEA), an endogenous lipid with anti-inflammatory and neuroprotective properties, has emerged as a promising therapeutic agent in managing AD. This study investigated the therapeutic effects of chronic (6-months) PEA administration via subcutaneous pellet in Tg2576 mice, a validated model of AD. The impact of PEA on amyloid precursor protein (APP) processing, astrocytic activation, microglial reactivity and neuroinflammation, nitrosative stress, dendritic spine density in hippocampal CA1 pyramidal neurons, and cognitive performance was assessed. Chronic PEA treatment of Tg2576 mice increased the expression of the α-secretase ADAM9 and reduced astrogliosis. Furthermore, PEA attenuated microglia reactivity, downregulated pro-inflammatory (CXCL13, MCP-1, GCSF) and upregulated anti-inflammatory (CXC3CL1 and IL-9) cytokine expression. Chronic PEA administration also decreased protein nitrosylation, downregulated calcineurin expression, restored dendritic spine density, and improved cognitive functions. Chronic PEA administration offers a promising therapeutic approach for AD by mitigating neuroinflammation, oxidative stress, and synaptic dysfunction, ultimately leading to cognitive function restoration.
Collapse
Affiliation(s)
- Daniel Tortolani
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Davide Decandia
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Giacomo Giacovazzo
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Lucia Scipioni
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Anna Panuccio
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | | | - Fabiola Eugelio
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Federico Fanti
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | | | - Livia La Barbera
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Debora Cutuli
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Dario Compagnone
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Marcello D’Amelio
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Roberto Coccurello
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Institute for Complex Systems (ISC), National Council of Research (CNR), Rome, Italy
| | - Sergio Oddi
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Laura Petrosini
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
| | - Mauro Maccarrone
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
144
|
Hamzavi SF, Elahi Vahed I, Samadi Shams A, Nozari F, Gamzeh Latava B, Mardukhi S, Sabaghi B, Hosseini ZS, Masoumi Shahr-e Babak Z, Ahrari S, Keshavarzian A, Rahmanian M. Association between polychlorinated biphenyls and hypertension risk: a systematic review and meta-analysis. Front Cardiovasc Med 2025; 12:1529431. [PMID: 40313580 PMCID: PMC12043693 DOI: 10.3389/fcvm.2025.1529431] [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: 11/16/2024] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Background and Aim Hypertension (HTN) is a widespread global health challenge, and its increasing prevalence is attributed to individual and environmental risk factors. Persistent organic pollutants (POPs), especially polychlorinated biphenyls (PCBs), contribute to cardiovascular risk by accumulating in fatty tissues, which leads to oxidative stress and vascular inflammation. This review and meta-analysis aimed to investigate the association between PCB exposure and hypertension. Methods Adhering to the PRISMA 2020 guidelines, data sources such as PubMed, Scopus, Web of Science, and Google Scholar were systematically searched up to July 2024 to find observational studies on the link between PCBs and hypertension risk. Studies were reviewed and chosen according to established inclusion and exclusion criteria, focusing on observational studies examining PCB exposure and hypertension risk. Independent reviewers conducted data extraction, and the quality of studies was evaluated using the JBI critical appraisal tool. A meta-analysis with a random-effects model was conducted to determine combined odds ratios (ORs) for hypertension linked to total PCB exposure and specific PCB types. Results Of the 494 records identified, 21 studies met the inclusion criteria, comprising 5 cohort studies, 15 cross-sectional studies, and one case-control study, totaling 51,514 participants. Exposure to total PCBs correlated with an elevated risk of hypertension (OR = 1.78, 95% CI: 1.30-2.44). Dioxin-like PCBs were also associated with a heightened risk (OR = 1.54, 95% CI: 1.24-1.90), while non-dioxin-like PCBs were not significantly linked (OR = 1.16, 95% CI: 0.81-1.66). Among individual congeners, PCB-74, PCB-118, PCB-105, and PCB-153 were significantly related to higher hypertension risk. Conclusion These findings indicate a positive correlation between PCB exposure and hypertension, particularly with dioxin-like PCBs and certain PCB congeners. Additional research is necessary to clarify the mechanisms involved and to promote measures for reducing PCB exposure, particularly in high-risk populations. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42024595223, PROSPERO (CRD42024595223).
Collapse
Affiliation(s)
- Seyedeh Fatemeh Hamzavi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Iman Elahi Vahed
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Fateme Nozari
- Student Research Committee, Tehran University of Medical Sciences, Tehran, Iran
| | - Baroukh Gamzeh Latava
- School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Hormozgan, Iran
| | - Saman Mardukhi
- Department of Occupational Health and Safety, School of Public Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behnoosh Sabaghi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zakieh Sadat Hosseini
- Department of Public Health, Faculty of Health and Paramedicine, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | | | - Sahar Ahrari
- Pharmaceutical Sciences Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Keshavarzian
- School of Medicine, Golestan University of Medical Sciences, Gorgan, Golestan, Iran
| | - Mohammad Rahmanian
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
145
|
Hu X, Dou Q, Jiang P, Zhang M, Wang J. Targeting matrix metalloproteinases activating and indoleamine 2,3-dioxygenase suppression for triple-negative breast cancer multimodal therapy. Int J Biol Macromol 2025; 310:143289. [PMID: 40253020 DOI: 10.1016/j.ijbiomac.2025.143289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
The dense extracellular matrix (ECM) and immunosuppressive tumor microenvironment represent two major challenges in the treatment of triple-negative breast cancer (TNBC). To address these obstacles, this study has developed a polymer micelle (NTP) for ECM remodeling and mitigation the immune microenvironment, based on activating endogenous matrix metalloproteinases (MMP) and suppression indoleamine 2,3-dioxygenase (IDO). Through self-assembly technology, this micelle effectively incorporates chemotherapy drugs (camptothecin (CPT) and cinnamaldehyde (CA)), reactive oxygen species (ROS) stimulants, nitric oxide (NO) donor and IDO inhibitor (NLG919), where CPT and CA have been reported to help generating ROS mainly in the mitochondrion. The guanidine group of poly-L-arginine (PArg), as an NO donor, can react with ROS to generate NO. The micelles aim to achieve significant therapeutic outcomes through robust drug penetration and anti-tumor immunity in multimodal therapy. They exhibit remarkable tumor tissue penetration ability, facilitating precise targeting of mitochondria and ROS production stimulation. Building upon this therapeutic foundation, the micellar system achieves in situ NO release, which effectively degrades the ECM through the activation of MMPs, while simultaneously promoting tumor cells apoptosis. Furthermore, the encapsulated NLG919 can be released and effectively mitigating the immunosuppressive milieu and triggering anti-tumor immune responses. Experimental results demonstrate that the micelles exhibit significant anti-tumor effects both in vitro and in vivo, accompanied by favorable biocompatibility. This study provides new insights into the application of subcellular targeting drug delivery systems in TNBC treatment, potentially heralding a new breakthrough in TNBC therapy.
Collapse
Affiliation(s)
- Xiaoxiao Hu
- School of Pharmacy, National Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China; Pharmaceutical Department, Baoding Second Hospital, Baoding 071051, China
| | - Qingqing Dou
- School of Pharmacy, National Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Peixiao Jiang
- School of Pharmacy, National Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Mo Zhang
- School of Pharmacy, National Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China.
| | - Jing Wang
- School of Pharmacy, National Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
146
|
Ozhan O, Ermis N, Celbis O, Samdanci E, Petekkaya S, Oruc M, Soylu O, Koparir P, Acet A, Parlakpinar H. Acute and subacute cardiovascular effects of synthetic cannabinoid JWH-018 in rat. Forensic Toxicol 2025:10.1007/s11419-025-00720-9. [PMID: 40240703 DOI: 10.1007/s11419-025-00720-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
PURPOSE This study investigates the cardiovascular effects of the synthetic cannabinoid naphthalene-1-yl-(1-pentylindole-3-yl)methanone (JWH-018) in rats. The research aims to evaluate the pharmacologic, cardiologic, biochemical, and histopathological effects of acute and subacute administration at low and high doses. The primary research question is how JWH-018 impacts heart function, blood pressure, ECG patterns, and cardiac tissue integrity. METHODS Wistar albino rats were divided into five groups: control, acute low-dose (ALD, 0.5 mg/kg), acute high-dose (AHD, 5 mg/kg), subacute low-dose (SALD, 0.5 mg/kg for 14 days), and subacute high-dose (SAHD, 5 mg/kg for 14 days). Cardiovascular effects were assessed using echocardiography, hemodynamic and ECG analysis, histopathology, biochemical markers, and LC-MS/MS quantification of JWH-018 and its metabolites in heart tissue. RESULTS Acute high-dose JWH-018 caused bradycardia and hypotension, while subacute high-dose increased heart rate but continued to lower blood pressure. JWH-018 induced cardiac arrhythmias, conduction blocks, and ischemic ECG changes, with prolonged QT intervals in subacute high-dose rats. Histopathological findings revealed myocardial infarction-like features, including contraction bands and ischemic damage, particularly in subacute groups. Elevated pro-BNP and triglycerides indicated cardiac stress and metabolic effects. JWH-018 and its metabolites were detected in heart tissue, primarily in high-dose groups. CONCLUSIONS JWH-018 has significant cardiovascular risks, causing heart rate dysregulation, hypotension, arrhythmias, and ischemic damage. These effects depend on dose and duration. The study highlights the potential dangers of synthetic cannabinoids, emphasizing that they should not be considered safe alternatives to natural cannabis.
Collapse
Affiliation(s)
- Onural Ozhan
- Department of Pharmacology, Faculty of Medicine, Inonu University, 44280, Malatya, Türkiye.
| | - Necip Ermis
- Department of Cardiology, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Osman Celbis
- Department of Forensic Medicine, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Emine Samdanci
- Department of Pathology, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Semih Petekkaya
- Department of Forensic Medicine, Faculty of Medicine, Canakkale Onsekiz Mart University, Canakkale, Türkiye
| | - Mucahit Oruc
- Department of Forensic Medicine, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Ozcan Soylu
- Department of Chemistry, Forensic Medicine Institute, Malatya, Türkiye
| | - Pelin Koparir
- Department of Chemistry, Forensic Medicine Institute, Malatya, Türkiye
| | - Ahmet Acet
- Department of Pharmacology, Faculty of Medicine, Inonu University, 44280, Malatya, Türkiye
| | - Hakan Parlakpinar
- Department of Pharmacology, Faculty of Medicine, Inonu University, 44280, Malatya, Türkiye
| |
Collapse
|
147
|
Ahmed HS. Neuropharmacological effects of calycosin: a translational review of molecular mechanisms and therapeutic applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04154-3. [PMID: 40237798 DOI: 10.1007/s00210-025-04154-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Calycosin, a naturally occurring isoflavonoid found predominantly in Astragalus membranaceus, exhibits significant therapeutic potential in various neurological conditions. Its multifaceted bioactive properties-antioxidant, anti-inflammatory, and anti-apoptotic-position it as a promising candidate for neuroprotection and neuroregeneration. This review explores calycosin's mechanisms of action, including its modulation of key signaling pathways such as HMGB1/TLR4/NF-κB (high mobility group box 1/toll-like receptor 4/nuclear factor kappa B), phosphatidylinositol-3-kinase (PI3 K)/Akt, ERK1/2 (extracellular signal-regulated kinase 1/2), and Hsp90/Akt/p38. In cerebral ischemia/reperfusion injury, calycosin reduces oxidative stress markers like ROS (reactive oxygen species) and MDA (malondialdehyde), enhances antioxidant enzymes (SOD (superoxide dismutase) and GPX (glutathione peroxidase)), and downregulates pro-inflammatory cytokines (TNF-α, IL-1β) through the HMGB1/TLR4/NF-κB pathway. It also inhibits autophagy via the STAT3/FOXO3a pathway and apoptosis by modulating Bax and Bcl-2 expression. In neuro-oncology, calycosin inhibits glioblastoma cell migration and invasion by modulating the TGF-β-mediated mesenchymal properties and suppressing the c-Met and CXCL10 signaling pathways. Additionally, it enhances the efficacy of temozolomide in glioma treatment through apoptotic pathways involving caspase-3 and caspase-9. Calycosin shows promise in Alzheimer's disease by reducing β-amyloid production and tau hyperphosphorylation via the GSK-3β pathway and improving mitochondrial function through the peroxisome proliferator-activated receptor gamma coactivator 1-Alpha (PGC-1α)/mitochondrial transcription factor A (TFAM) signaling pathway. In Parkinson's disease, calycosin mitigates oxidative stress, prevents dopaminergic neuronal death, and reduces neuroinflammation by inhibiting the TLR/NF-κB and MAPK pathways. It has also shown therapeutic potential in meningitis and even neuroprotective effects against hyperbilirubinemia-induced nerve injury. Despite these promising findings, further research, including detailed mechanistic studies and clinical trials, is needed to fully understand calycosin's therapeutic mechanisms and validate its potential in human subjects. Developing advanced delivery systems and exploring synergistic therapeutic strategies could further enhance its clinical application and effectiveness.
Collapse
Affiliation(s)
- H Shafeeq Ahmed
- Bangalore Medical College and Research Institute, K.R Road, Bangalore, 560002, Karnataka, India.
| |
Collapse
|
148
|
Jaruga P, Kant M, Luzadder MM, Lloyd RS, Boldogh I, Dizdaroglu M. Inhibition by 4-(4-Bromo-2-oxo-3 H-benzimidazol-1-yl)- N-(4-iodophenyl)piperidine-1-carboxamide (TH5487) of the Activity of Human 8-Oxoguanine DNA Glycosylase-1 (OGG1) for the Excision of 2,6-Diamino-4-hydroxy-5-formamidopyrimidine, 4,6-Diamino-5-formamidopyrimidine, and 8-Oxoguanine from Oxidatively Damaged DNA. Biochemistry 2025; 64:1788-1796. [PMID: 40179276 PMCID: PMC12004446 DOI: 10.1021/acs.biochem.4c00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025]
Abstract
DNA glycosylases of the base excision repair pathway have become clinically validated drug targets for the treatment of several diseases. Human OGG1 (hOGG1) is specific for the removal of the highly mutagenic 8-oxoguanine (8-oxo-Gua) and 2,6-diamino-4-hydroxy-5-formamidopyrimidine (FapyGua) from damaged DNA. To develop clinically approved drugs, various small-molecule inhibitors of hOGG1 have been developed to inhibit its glycosylase and lyase activities, with 4-(4-bromo-2-oxo-3H-benzimidazol-1-yl)-N-(4-iodophenyl)piperidine-1-carboxamide (TH5487) shown to be a potent inhibitor. The inhibition of hOGG1 by TH5487 has been shown to suppress cancer cell growth, pulmonary inflammation, and lung fibrosis and sensitize cancer cells to ionizing radiation, confirming hOGG1 as a target for pharmaceutical intervention. While the assays that identified TH5487 utilized an oligodeoxynucleotide with the target substrate being 8-hydroxyadenine mispaired with cytosine, measurements of TH5487-mediated inhibition of the release of 8-oxo-Gua and FapyGua have not been reported. In the present work, we investigated the inhibition of hOGG1 by TH5487 using genomic DNA with multiple lesions and gas chromatography-tandem mass spectrometry with isotope dilution to measure inhibition of hOGG1-catalyzed DNA base lesion removal from DNA. An oligodeoxynucleotide containing 8-oxo-Gua was also used to measure the half-maximal inhibitory concentration (IC50), which is 0.800 μmol/L ± 0.061 μmol/L. We show that TH5487 efficiently inhibits the excision of both 8-oxo-Gua and FapyGua, and a minor substrate 4,6-diamino-5-formamidopyrimidine (FapyAde) from DNA with the IC50 values of 1.6 μmol/L, 3.1 μmol/L, and 3.1 μmol/L, respectively. The results suggest that the approach used in the present work may be applied for future studies of hOGG1 inhibition by TH5487 on cellular and animal disease models.
Collapse
Affiliation(s)
- Pawel Jaruga
- Biomolecular
Measurement Division, National Institute
of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Melis Kant
- Biomolecular
Measurement Division, National Institute
of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Michael M. Luzadder
- Oregon
Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon 97239, United States
| | - R. Stephen Lloyd
- Oregon
Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon 97239, United States
- Department
of Molecular and Medical Genetics, Oregon
Health & Science University, Portland, Oregon 97239, United States
| | - Istvan Boldogh
- Department
of Microbiology and Immunology, University
of Texas Medical Branch at Galveston, Galveston, Texas 77555, United States
| | - Miral Dizdaroglu
- Biomolecular
Measurement Division, National Institute
of Standards and Technology, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
149
|
Arner P. Signalling from adipose tissue to the brain is not just about leptin. Nat Metab 2025:10.1038/s42255-025-01277-y. [PMID: 40234626 DOI: 10.1038/s42255-025-01277-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Affiliation(s)
- Peter Arner
- Department of Medicine, Karolinska Institutet, C2-94, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
150
|
Hu M, Oliveira APBN, Fang Z, Feng Y, Miranda M, Kowli S, Arunachalam PS, Vasudevan G, Hui HSY, Grifoni A, Sette A, Litvack M, Rouphael N, Suthar MS, Ji X, Maecker HT, Hagan T, Dhillon G, Nicolls MR, Pulendran B. Altered baseline immunological state and impaired immune response to SARS-CoV-2 mRNA vaccination in lung transplant recipients. Cell Rep Med 2025; 6:102050. [PMID: 40187358 PMCID: PMC12047491 DOI: 10.1016/j.xcrm.2025.102050] [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: 08/28/2024] [Revised: 12/08/2024] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
The effectiveness of COVID-19 mRNA vaccines is diminished in organ transplant patients. Using a multi-omics approach, we investigate the immunological state of lung transplant (LTX) recipients at baseline and after SARS-CoV-2 mRNA vaccination compared to healthy controls (HCs). LTX patients exhibit a baseline immune profile resembling severe COVID-19 and sepsis, characterized by elevated pro-inflammatory cytokines (e.g., EN-RAGE [also known as S100A12], interleukin [IL]-6), reduced human leukocyte antigen (HLA)-DR expression on monocytes and dendritic cells, impaired cytokine production, and increased plasma microbial products. Single-cell RNA sequencing identifies an enriched monocyte cluster in LTX patients marked by high S100A family expression and reduced cytokine and antigen presentation genes. Post vaccination, LTX patients show diminished antibody, B cell, and T cell responses, along with blunted innate immune signatures. Integrative analysis links these altered baseline immunological features to impaired vaccine responses. These findings provide critical insights into the immunosuppressed condition of LTX recipients and their reduced vaccine-induced adaptive and innate immune responses.
Collapse
Affiliation(s)
- Mengyun Hu
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Ana Paula B N Oliveira
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zhuoqing Fang
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Yupeng Feng
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA; Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Molly Miranda
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Sangeeta Kowli
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA; Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Gowri Vasudevan
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Harold Sai-Yin Hui
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Matthew Litvack
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA, USA
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA, USA
| | - Mehul S Suthar
- Department of Pediatrics, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - Xuhuai Ji
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Thomas Hagan
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gundeep Dhillon
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Mark R Nicolls
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|