1
|
Lou S, Dai C, Wu Y, Wang L, Jin Y, Shen N, Lv W, Wu M, Xu X, Han J, Fan X. Betulonic acid: A review on its sources, biological activities, and molecular mechanisms. Eur J Pharmacol 2025; 998:177518. [PMID: 40107338 DOI: 10.1016/j.ejphar.2025.177518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Pentacyclic triterpenoids represent a significant class of phytochemicals, categorized into oleanane, ursane, friedelane, and lupane. Among these, betulonic acid stands out as a lupane-type pentacyclic triterpenoid found in numerous plants. Its diverse biological properties, including anti-tumor, anti-viral, anti-inflammatory, anti-bacterial, and hepato-protective effects, have been extensively documented. To further explore the therapeutic potential of betulonic acid and its derivatives, we provide a comprehensive review of their sources, biological activities, and molecular mechanisms. We aim for this synthesis of data to stimulate fresh perspectives on betulonic acid and its potential in drug discovery.
Collapse
Affiliation(s)
- Shengying Lou
- Department of Pathology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Chunyan Dai
- Department of Pathology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Yuhua Wu
- Department of Pathology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Lijiang Wang
- Department of Pathology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yuancheng Jin
- Department of Pathology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Naitao Shen
- Department of Pathology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wang Lv
- Department of Pathology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Miaolian Wu
- Department of Pathology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xiaojun Xu
- Department of Pathology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Center for Innovative Traditional Chinese Medicine Target and New Drug Research, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| | - Jichun Han
- College of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, China.
| | - Xiangcheng Fan
- Department of Pathology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Center for Innovative Traditional Chinese Medicine Target and New Drug Research, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
2
|
Yao F, Bao Y, Meng Q, Chen Y, Zhao L, Wang P, Zhou B. Periprosthetic osteolysis: Mechanisms and potential treatment strategies. Cell Signal 2025; 131:111758. [PMID: 40132773 DOI: 10.1016/j.cellsig.2025.111758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Periprosthetic osteolysis is a common bone-related disorder that often occurs after total hip arthroplasty. The implants can cause damage to bone and bone-related cells due to mechanical stress and micromotions, resulting in the generation of a large number of wear particles. These wear particles trigger inflammation and oxidative stress in the surrounding tissues, disrupting the delicate balance maintained by osteoblasts and osteoclasts, ultimately leading to bone loss around the implant. Clinical investigations have demonstrated that Epimedium prenylflavonoids, miR-19a-3p, stem cell-derived exosomes, and certain non-PPO category pharmaceuticals have regulatory effects on bone homeostasis through distinct molecular pathways. Notably, this phenomenon reflects inherent biological rationality rather than stochastic occurrence. Extensive research has revealed that multiple natural compounds, non-coding RNAs, exosomes, and non-PPO therapeutics not only exert modulatory influences on critical pathophysiological processes including inflammatory cascades, oxidative stress responses, and tissue regeneration mechanisms, but also effectively regulate bone-related cellular functions to inhibit PPO progression. Therefore, this review comprehensively and systematically summarizes the main pathogenic mechanisms of periprosthetic osteolysis. Furthermore, it delves deeper into the research progress on the applications of currently reported natural products, ncRNAs, exosomes, and non-PPO medications in the treatment of periprosthetic osteolysis. Based on this, we hope that this paper can provide new perspectives and references for the future development of drugs targeting periprosthetic osteolysis.
Collapse
Affiliation(s)
- Fang Yao
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yue Bao
- Department of Nursing, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Qian Meng
- Outpatient Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yanrong Chen
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Luxi Zhao
- Department of Anesthesiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Pingmei Wang
- Department of Orthopaedics, The People's Hospital of Shimen County, Shimen 415399, China
| | - Bin Zhou
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
3
|
Yang H, Xia Y, Ma Y, Gao M, Hou S, Xu S, Wang Y. Inhibition of the cGAS-STING pathway: contributing to the treatment of cerebral ischemia-reperfusion injury. Neural Regen Res 2025; 20:1900-1918. [PMID: 38993125 PMCID: PMC11691458 DOI: 10.4103/nrr.nrr-d-24-00015] [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: 01/05/2024] [Revised: 03/05/2024] [Accepted: 05/02/2024] [Indexed: 07/13/2024] Open
Abstract
The cGAS-STING pathway plays an important role in ischemia-reperfusion injury in the heart, liver, brain, and kidney, but its role and mechanisms in cerebral ischemia-reperfusion injury have not been systematically reviewed. Here, we outline the components of the cGAS-STING pathway and then analyze its role in autophagy, ferroptosis, cellular pyroptosis, disequilibrium of calcium homeostasis, inflammatory responses, disruption of the blood-brain barrier, microglia transformation, and complement system activation following cerebral ischemia-reperfusion injury. We further analyze the value of cGAS-STING pathway inhibitors in the treatment of cerebral ischemia-reperfusion injury and conclude that the pathway can regulate cerebral ischemia-reperfusion injury through multiple mechanisms. Inhibition of the cGAS-STING pathway may be helpful in the treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Hang Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yulei Xia
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yue Ma
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Mingtong Gao
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Shuai Hou
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Shanshan Xu
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Yanqiang Wang
- Department of Neurology II, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| |
Collapse
|
4
|
Chen Q, Zhong G, Fang X, Lin C, Wang S, Li M. The multifaceted role of Sestrin 3 (SESN3) in oxidative stress, inflammation and tumorigenesis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119938. [PMID: 40174866 DOI: 10.1016/j.bbamcr.2025.119938] [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: 02/19/2025] [Revised: 03/07/2025] [Accepted: 03/20/2025] [Indexed: 04/04/2025]
Abstract
The pathogenesis of inflammation and tumors is a focal point of scientific inquiry, with oxidative stress often serving as the primary initiator. Within the human genome, the SESN3 gene encodes the SESN3 protein, a crucial antioxidant stress protein. Acting as a regulatory factor, SESN3 intricately modulates cellular oxidative stress, actively participating in cellular protection and repair mechanisms. Its functions span antioxidative, anti-aging, and anti-tumor properties. The expression of SESN3 is closely linked to cellular and oxidative stress, metabolic status, and specific signaling pathways. This review aims to delve into the origins and functions of SESN3, its role within signaling pathways, and its contributions to inflammation and tumorigenesis.
Collapse
Affiliation(s)
- Qiusan Chen
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guoqiang Zhong
- Department of Gastroenterology, Guangzhou First People's Hospital, Guangzhou, Guangdong, China
| | - Xianmei Fang
- Department of Ultrasonography, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Chuangzhen Lin
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shanping Wang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mingsong Li
- Department of Gastroenterology, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
De Domenico S, La Banca V, D'Amico S, Nicolai S, Peschiaroli A. Defining the transcriptional routes controlling lncRNA NEAT1 expression: implications in cellular stress response, inflammation, and differentiation. Discov Oncol 2025; 16:768. [PMID: 40369379 PMCID: PMC12078918 DOI: 10.1007/s12672-025-02510-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Abstract
NEAT1 (Nuclear Enriched Abundant Transcript 1) is a long non-coding RNA playing a critical role in both physiological and pathological settings by directly modulating a variety of biological events, including transcriptional regulation, RNA processing, and chromatin remodeling. Multiple evidence demonstrated that different transcription factors and signaling pathways modulate biological processes by tightly regulating NEAT1 expression. These regulatory mechanisms act at different levels, allowing cells to rapidly modulate NEAT1 expression and dynamically respond to sudden changes in cellular conditions. In this review, we summarize and discuss the transcriptional routes controlling NEAT1 expression, emphasizing recent evidence showing the pivotal role of NEAT1 in regulating important biological processes, such as cellular stress response, inflammation, and cell differentiation.
Collapse
Affiliation(s)
- Sara De Domenico
- Department of Experimental Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Veronica La Banca
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133, Rome, Italy
| | - Silvia D'Amico
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133, Rome, Italy
| | - Sara Nicolai
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133, Rome, Italy.
| | - Angelo Peschiaroli
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133, Rome, Italy.
| |
Collapse
|
6
|
Kim MW, Kipnis J. Glymphatics and meningeal lymphatics unlock the brain-immune code. Immunity 2025; 58:1040-1051. [PMID: 40324376 DOI: 10.1016/j.immuni.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 05/07/2025]
Abstract
The central nervous system (CNS) was once perceived as entirely shielded from the immune system, protected behind the blood-brain barrier and thought to lack lymphatic drainage. However, recent evidence has challenged many dogmas in neuroimmunology. Indeed, by means of glymphatics, brain-derived "waste" from deep within the CNS mobilizes toward immunologically active brain borders, where meningeal lymphatic vessels are appropriately positioned to drain antigens from the brain to the periphery. Accordingly, the presentation of brain-derived self-peptides emerges at the brain's borders and drives T cell responses with suppressive properties, critical in allowing active immunosurveillance while limiting aberrant immune reactivity. Taking into consideration these concepts, we further discuss how inflammation, aging, and neurodegenerative diseases potentially reshape the repertoire of self-antigens and immune cells, disrupting the healthy dialogue between the CNS and immune system. Collectively, this evolving perspective unveils new therapeutic avenues for CNS pathologies.
Collapse
Affiliation(s)
- Min Woo Kim
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Medical Scientist Training Program, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Medical Scientist Training Program, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Silveira LL, Santos Dias MMD, Pelinsari SM, Paula RAD, Castro ASB, Almeida VLD, Gonçalves RV. Trichilia silvatica extracts modulate the oxinflammatory response: an in vitro analysis. JOURNAL OF ETHNOPHARMACOLOGY 2025:119973. [PMID: 40368256 DOI: 10.1016/j.jep.2025.119973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 05/01/2025] [Accepted: 05/12/2025] [Indexed: 05/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Plants belonging to the Meliaceae family, such as Trichilia silvatica C. DC., known as catiguá-branco, have attracted considerable interest in phytochemical research due to their diverse and significant secondary metabolites. Trichilia silvatica has traditionally been employed in Brazilian medicine to treat inflammatory disorders. Moreover, studies have reported its antioxidant and antimicrobial properties, highlighting its potential therapeutic applications. AIM OF THE STUDY This study aimed to evaluate the potential of Trichilia silvatica leaf and stem extracts in modulating OxInflammation in RAW264.7 macrophage cells following exposure to lipopolysaccharide (LPS) or hydrogen peroxide (H2O2) and to elucidate the underlying mechanisms of action. MATERIAL AND METHODS The phytochemical composition of the extracts was characterized using thin-layer chromatography (TLC), HPLC equipped with a reversed-phase Hypersil C-18 column, and spectrophotometric method. Their antioxidant activity was evaluated using the 2,2-difenil-1-picrilhidrazil (DPPH) and Ferric Reducing Antioxidant Power (FRAP) assays. Cell viability was assessed via the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, alongside the determination of catalase (CAT) and superoxide dismutase (SOD) enzyme activities, as well as nitric oxide (NO) production in cells treated with the extracts and subsequently stimulated with H2O2. Gene expression levels of Factor nuclear kappa B (NF-κB), Ciclooxygenase 2 (COX-2), Tumor necrosis factor alpha (TNF-α), Interleukin 10 (IL-10), and Hypoxia-inducible factor-1 (HIF-1) were quantified using RT-qPCR. RESULTS Trichilia silvatica extracts revealed the presence of terpenes/steroids, coumarins, condensed tannins, and phenolic acids, including chlorogenic and caffeic acids. The findings indicate that the leaf extract at 100 μg/ml and the stem extract at 100 μg/ml and 250 μg/ml preserved or enhanced cell viability, conferring protection against H2O2-induced oxidative stress. These concentrations significantly increased CAT activity, whereas SOD activity remained unaffected. Nitric oxide production was significantly reduced when cells were treated with 100 μg/ml and 250 μg/ml of both leaf and stem extracts. Moreover, FRAP value revealed an increase in antioxidant capacity at 250 μg/mL. Both leaf and stem extracts, at 100 μg/mL and 250 μg/mL, exhibited a DPPH radical scavenging capacity exceeding 75 % and downregulated the expression of pro-inflammatory cytokines, including NF-κB, TNF-α, and COX-2. Notably, the leaf extract at 250 μg/ml and the stem extract at 100 μg/mL upregulated the expression of IL-10 and H1F1. CONCLUSIONS These findings indicate that Trichilia silvatica extracts exhibit notable antioxidant activity, as evidenced by greater than 75% inhibition of DPPH radicals and elevated FRAP values. Additionally, the extracts demonstrated anti-inflammatory properties by downregulating key pro-inflammatory mediators, including TNF-α, NF-κB, and COX-2, while upregulating the anti-inflammatory cytoline IL-10 and enhancing enhancing tissue oxygenation and nutrient supply through increased expression of HIF-1. These effects highlight the potential of T. silvatica extracts as therapeutic agents for managing inflammatory diseases associated with oxidative stress, thereby supporting their traditional medicinal use.
Collapse
Affiliation(s)
- Leonardo Lopes Silveira
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | | | - Silvânia Mól Pelinsari
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | | | | | - Vera Lúcia de Almeida
- Phytochemistry and Pharmaceutical Prospecting Service, Division of Science and Innovation, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil.
| | - Reggiani Vilela Gonçalves
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais 36570-900, Brazil; Plants for Human Health Institute, North Carolina Research Campus, 600 Laureate Way, 28081 Kannapolis, NC.
| |
Collapse
|
8
|
Dai D, Zhang Z, Ma M, Zhao C, Li J, Zhang S, Ma P, Wu Q, Song D. Low-background Near-infrared Fluorescent Probe for Real-time Monitoring of β-Glucuronidase Activity in Inflammation and Therapy. Anal Chem 2025; 97:9414-9421. [PMID: 40272894 DOI: 10.1021/acs.analchem.5c00658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
β-Glucuronidase (GUS) is an acidic hydrolase enzyme overexpressed in various inflammatory diseases, making it a promising biomarker for inflammation. However, current tools for real-time, in situ imaging of GUS activity are hindered by background interference, which reduces their effectiveness in dynamic biological environments. To address this challenge, we developed Ox-GUS, a GUS-specific fluorescent probe with a unique molecular design featuring a disrupted conjugated structure. This design provided Ox-GUS with near-zero background optical properties, a significantly enhanced signal-to-noise ratio, and a highly sensitive detection ability. The probe demonstrated a fluorescence enhancement of up to 400 folds in response to GUS activity, with a detection limit as low as 0.0035 U/mL. We successfully employed Ox-GUS to visualize GUS activity in real-time in mouse models of rheumatoid arthritis, autoimmune hepatitis, and inflammatory bowel disease, and effectively monitored therapeutic responses. This study highlights the potential of Ox-GUS as a robust tool for advancing research on GUS-related inflammatory mechanisms and for early diagnosis and treatment monitoring of inflammatory diseases.
Collapse
Affiliation(s)
- Dianfeng Dai
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Zhimin Zhang
- Department of Pharmacy, Changchun Medical College, Changchun 130031, China
| | - Mo Ma
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
- School of Pharmacy, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Chen Zhao
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Jingkang Li
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Siqi Zhang
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Pinyi Ma
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Qiong Wu
- Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, The Third Bethune Hospital of Jilin University, Sendai Street 126, Changchun 130033, China
| | - Daqian Song
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| |
Collapse
|
9
|
Wu YJ, Li SM, Chen CL, Chen ZR, Chen JJ. Anti-inflammatory activity of Pogostemon cablin: Bioactive components and their modulation of MAPK and NF-κB signaling pathway. Bioorg Chem 2025; 161:108516. [PMID: 40345124 DOI: 10.1016/j.bioorg.2025.108516] [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: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025]
Abstract
Pogostemon cablin is a well-known Lamiaceae plant and widely utilized in Traditional Chinese Medicine (TCM) for its neuroprotective, anti-inflammatory, and anxiolytic properties. In this study, 16 known compounds (1-15 and 17) and one semi-synthesized new compound, 5-hydroxy-3-isoprenyloxy-7,3',4'-trimethoxyflavone (16), including flavonoids, pyranones, sesquiterpenes, and benzenoids, were obtained and characterized from aerial parts of P. cablin and investigated for their anti-inflammatory properties through the MAPK and NF-κB signaling pathways in LPS-induced RAW264.7 macrophages. Among the isolated compounds, rhamnazin (4), pachypodol (5), and (E)-2-methyl-6-(p-tolyl)hept-3-en-2-ol (15) exhibited potent anti-inflammatory activities in LPS-induced RAW264.7 macrophages. Rhamnazin (4) significantly modulated IκBα levels and reduced the expressions of phosphorylation of JNK and p38, indicating its effects on suppressing NF-κB activation and mitigating inflammation via MAPK signaling. Pachypodol (5) selectively inhibited iNOS and p-JNK expressions, showing specificity in its anti-inflammatory activity. (E)-2-Methyl-6-(p-tolyl)hept-3-en-2-ol (15) downregulated iNOS, p-Erk, and p-JNK expressions, demonstrating a broader inhibitory profile on pro-inflammatory mediators. Further molecular docking results demonstrated bioactive compounds 4, 5, and 15 possessed strong binding affinities with key residues, particularly Hem901, Pro344, and Glu371, consistent with their NO inhibition effects. In addition, in silico prediction of physicochemical properties confirmed favorable oral bioavailability and drug-likeness, supporting their potential as lead compounds for anti-inflammatory drug development. These findings provide comprehensive molecular insight into the anti-inflammatory effects and reveal the therapeutic potential of P. cablin constituents as natural plant-derived NF-κB and MAPK-targeting anti-inflammatory agents, offering promising candidates for managing inflammatory diseases.
Collapse
Affiliation(s)
- Yu-Jing Wu
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Sin-Min Li
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Zih-Rong Chen
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Jih-Jung Chen
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404333, Taiwan; Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan.
| |
Collapse
|
10
|
Lütge M, Kurz L, Stanossek Y, Meili S, Cheng HW, De Martin A, Brandstadter J, Maillard I, Robinson MD, Stoeckli SJ, Pikor NB, Onder L, Ludewig B. Fibroblastic reticular cells form reactive myeloid cell niches in human lymph nodes. Sci Immunol 2025; 10:eads6820. [PMID: 40315298 DOI: 10.1126/sciimmunol.ads6820] [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: 08/23/2024] [Accepted: 04/07/2025] [Indexed: 05/04/2025]
Abstract
Lymph nodes play a key role in maintaining fluid balance in homeostatic and inflamed tissues and provide fibroblastic niche environments for optimal immune cell positioning and interaction. Here, we used single-cell and spatial transcriptomic analyses in combination with high-resolution imaging to molecularly define and functionally characterize niche-forming cells that control inflammation-driven remodeling in human lymph nodes. Fibroblastic reticular cells responded to inflammatory perturbation with activation and expansion of poised niche environments. Inflammation-induced adaptation of lymph node infrastructure and topography included the expansion of peptidase inhibitor 16 (PI16)-expressing reticular cell (PI16+ RC) networks that enwrap the perivenular conduit system. Interactome analyses indicated that macrophage-derived oncostatin M directs PI16+ RC activation in inflamed lymph nodes and thereby promotes immune cell aggregation in the perivenular space. In conclusion, these data demonstrate that the inflammatory remodeling of human lymph nodes results in the formation of reactive myeloid cell niches by PI16+ RCs.
Collapse
Affiliation(s)
- Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Lisa Kurz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Yves Stanossek
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Samuel Meili
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Joshua Brandstadter
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark D Robinson
- Department of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Sandro J Stoeckli
- Department of Otorhinolaryngology, Head and Neck Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Natalia B Pikor
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Institute of Microbiology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- University Heart Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Jin J, Yue L, Du M, Geng F, Gao X, Zhou Y, Lu Q, Pan X. Molecular Hydrogen Therapy: Mechanisms, Delivery Methods, Preventive, and Therapeutic Application. MedComm (Beijing) 2025; 6:e70194. [PMID: 40297245 PMCID: PMC12035766 DOI: 10.1002/mco2.70194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Molecular hydrogen (H2), recognized as the smallest gas molecule, is capable of permeating cellular membranes and diffusing throughout the body. Due to its high bioavailability, H2 is considered a therapeutic gas for the treatment of various diseases. The therapeutic efficacy of hydrogen is contingent upon factors such as the administration method, duration of contact with diseased tissue, and concentration at targeted sites. H2 can be administered exogenously and is also produced endogenously within the intestinal tract. A comprehensive understanding of its delivery mechanisms and modes of action is crucial for advancing hydrogen medicine. This review highlights H₂'s mechanisms of action, summarizes its administration methods, and explores advancements in treating intestinal diseases (e.g., inflammatory bowel disease, intestinal ischemia-reperfusion, colorectal cancer). Additionally, its applications in managing other diseases are discussed. Finally, the challenges associated with its clinical application and potential solutions are explored. We propose that current delivery challenges faced by H2 can be effectively addressed through the use of nanoplatforms; furthermore, interactions between hydrogen and gut microbiota may provide insights into its mechanisms for treating intestinal diseases. Future research should explore the synergistic effects of H2 in conjunction with conventional therapies and develop personalized treatment plans to achieve precision medicine.
Collapse
Affiliation(s)
- Jiayi Jin
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Lijun Yue
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Maoru Du
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Feng Geng
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Xue Gao
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Yuming Zhou
- Department of Laboratory MedicineYantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Qianqian Lu
- Department of OncologyYantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Xiaohong Pan
- School of PharmacyBinzhou Medical UniversityYantaiChina
| |
Collapse
|
12
|
Goldoni FC, Benvenutti L, Nunes R, Vaz CR, Garcia L, Furtado K, Dos Santos Bubniak L, de Campos Buzzi F, Corrêa R, Quintão NLM, Santin JR. Safety evaluation and modulatory effects on innate immune system of pyrazoline-derived compounds. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5677-5691. [PMID: 39601822 DOI: 10.1007/s00210-024-03653-z] [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: 06/04/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Pyrazolines are compounds that have been studied for their strong biological potential and structure diversity. Several studies demonstrated their biological effectiveness, highlighting their anti-inflammatory potential. This study aimed to evaluate the physicochemical profile, the safety, and the anti-inflammatory effects of four pyrazolines (PH0, PH3, PH4, and PH7). Initially, in silico analysis were performed on SwissADME and QSAR Toolbox platforms. The anti-inflammatory activity was assessed by in vitro and in vivo methodologies. Neutrophils collected from mice peritoneum and macrophages immortalized cell line (Raw 264.7) were stimulated with lipopolysaccharide (LPS), and subsequent measurement of nitric oxide (NO) and IL-1β, TNF, and IL-6 cytokines were performed by ELISA method. The effect on cell migration was evaluated by chemotaxis assay. The effect on efferocytosis was investigated using senescent neutrophils and macrophages from mice's bone marrow. The in silico results suggest suitable properties for a pharmacological prototype for oral administration, with no significant toxic effects. All compounds significantly reduced NO levels, as well as levels of IL-1β, TNF, and IL-6 cytokines. Also, they were able to reduce cell migration and increase efferocytosis. The in vivo air pouch model confirmed the effects of pyrazolines on cell kinetics and on the levels of cytokines (IL-1β and TNF) on the air pouch lavage. All of the pyrazolines evaluated showed to have positive effects on mechanisms that modulate the inflammatory response. Furthermore, the in silico analysis suggests that chemical changes in the structure can lead to improvement of the biological and pharmacokinetics proprieties.
Collapse
Affiliation(s)
- Fernanda Capitanio Goldoni
- Postgraduate Program in Pharmaceutical Science, Universidade Do Vale Do Itajaí (UNIVALI), 458, Bloco F6, ECS, Sala 316, CEP, Itajaí, SC, 88302-901, Brazil
| | - Larissa Benvenutti
- Postgraduate Program in Pharmaceutical Science, Universidade Do Vale Do Itajaí (UNIVALI), 458, Bloco F6, ECS, Sala 316, CEP, Itajaí, SC, 88302-901, Brazil
| | - Roberta Nunes
- Postgraduate Program in Pharmaceutical Science, Universidade Do Vale Do Itajaí (UNIVALI), 458, Bloco F6, ECS, Sala 316, CEP, Itajaí, SC, 88302-901, Brazil
| | - Carlos Rafael Vaz
- Postgraduate Program in Pharmaceutical Science, Universidade Do Vale Do Itajaí (UNIVALI), 458, Bloco F6, ECS, Sala 316, CEP, Itajaí, SC, 88302-901, Brazil
| | - Louise Garcia
- Pharmacy Course, School of Health Sciences, Universidade Do Vale Do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Keyla Furtado
- Pharmacy Course, School of Health Sciences, Universidade Do Vale Do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Lorena Dos Santos Bubniak
- Pharmacy Course, School of Health Sciences, Universidade Do Vale Do Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Fátima de Campos Buzzi
- Postgraduate Program in Pharmaceutical Science, Universidade Do Vale Do Itajaí (UNIVALI), 458, Bloco F6, ECS, Sala 316, CEP, Itajaí, SC, 88302-901, Brazil
| | - Rogério Corrêa
- Postgraduate Program in Pharmaceutical Science, Universidade Do Vale Do Itajaí (UNIVALI), 458, Bloco F6, ECS, Sala 316, CEP, Itajaí, SC, 88302-901, Brazil
| | - Nara Lins Meira Quintão
- Postgraduate Program in Pharmaceutical Science, Universidade Do Vale Do Itajaí (UNIVALI), 458, Bloco F6, ECS, Sala 316, CEP, Itajaí, SC, 88302-901, Brazil
| | - José Roberto Santin
- Postgraduate Program in Pharmaceutical Science, Universidade Do Vale Do Itajaí (UNIVALI), 458, Bloco F6, ECS, Sala 316, CEP, Itajaí, SC, 88302-901, Brazil.
| |
Collapse
|
13
|
Cao Z, Yao Y, Cai M, Zhang C, Liu Y, Xin H, An B, Wang H, Lu Y, Li Z, Chen Y, Huang Y, Xin M, Li R, Qian Z, Zhou Y, Xiang X, Moreau R, Xie Q. Blood markers for type-1, -2, and -3 inflammation are associated with severity of acutely decompensated cirrhosis. J Hepatol 2025; 82:836-850. [PMID: 39490592 DOI: 10.1016/j.jhep.2024.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/27/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND & AIMS In patients with acutely decompensated cirrhosis (ADC) who present with clinically apparent precipitants (i.e., infections, acute liver injury), alterations in blood markers of inflammation associate with progression toward severe phenotypes (e.g., acute-on-chronic liver failure [ACLF]). However, it is unclear whether alterations in blood inflammatory markers associate with progression of ADC independently of precipitants. METHODS We prospectively enrolled 394 patients admitted for ADC who were classified into four phenotypes of increasing severity: no organ dysfunction (n = 168), organ dysfunction alone (n = 72), organ failure without ACLF (n = 91), and ACLF (n = 63). Clinical blood cell counts and serum levels of inflammatory markers (including soluble markers related to type-1, type-2, and type-3 inflammation) were obtained at enrollment. Ordinal regression with adjacent categories logit model adjusted for confounders (including precipitants) was used to analyze associations between changes in each blood inflammatory marker and the worsening of ADC. RESULTS Inflammatory markers that were associated with a higher risk of progressing to the next more severe stage were as follows: increasing neutrophil counts (adjusted common odds ratio [cOR] 1.17, 95% CI 1.06-1.28); increasing levels of the type-2 cytokine interleukin (IL)-25 (cOR 1.21, 95% CI 1.06-1.39), type-3 cytokines IL-6 (cOR 1.15, 95% CI 1.02-1.28) and IL-22 (cOR 1.16, 95% CI 1.03-1.30), or anti-inflammatory soluble CD163 (cOR 1.94, 95% CI 1.58-2.38); decreasing lymphocyte counts (cOR 0.77, 95% CI 0.68-0.87); or decreasing levels of the type-1 cytokine IFN-γ (cOR 0.85, 95% CI 0.75-0.95). CONCLUSIONS Among patients with ADC, alterations in blood levels of cytokines related to type-1, type-2 and type-3 inflammation, together with neutrophilia, lymphopenia and elevated anti-inflammatory signals were individually associated with an increased risk of progressing toward ACLF, independently of the presence of clinically apparent precipitants. IMPACT AND IMPLICATIONS This study reveals that among patients with acutely decompensated cirrhosis, alterations in blood levels of cytokines related to type-1, type-2 and type-3 inflammation, together with neutrophilia, lymphopenia and elevated anti-inflammatory signals were individually associated with increased risk of progressing toward acute-on-chronic liver failure, independently of the presence of clinically apparent precipitants. These findings raise questions about the role of impaired barrier tissues and dysregulated production of blood immune cells in the pathophysiology of severe phenotypes of acutely decompensated cirrhosis, stimulating research to identify potential new biomarkers and targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- Zhujun Cao
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujing Yao
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Minghao Cai
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenxi Zhang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhan Liu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiguang Xin
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baoyan An
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yide Lu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziqiang Li
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaoxing Chen
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Huang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xin
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruokun Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuping Qian
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Nursing, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zhou
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaogang Xiang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Richard Moreau
- European Foundation for the Study of Chronic Liver Failure (EF CLIF), Barcelona, Spain; Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), Paris, France; Assistance Publique - Hôpitaux de Paris (APHP), Service d'Hépatologie, Hôpital, Beaujon, Clichy, France.
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Tuohudaali W, Ji TF, Ding WT, Li CY, Bianba JS, Ci R, Zhao J. Urolithin B inhibits LPS-induced macrophage M1 polarization via miR155-5p mediated MAPK/NF-кB pathway. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025; 27:787-796. [PMID: 39671344 DOI: 10.1080/10286020.2024.2435984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
This study investigated inhibiting mechanisms of Urolithin B (Uro B) on macrophage M1 polarization. Uro B (50 μM) could inhibit the PGE2, COX-2, NO, iNOS, TNF-α, IL-1β and IL-6 levels compared with model group (P < 0.05) as well as the CD86 and F4/80 expression. The miR155-5p overexpression could increase the p38 MAPK, JNK, ERK mRNA activities (P < 0.05), Uro B (50 μM) could reverse changes in these indicators (P < 0.05). Moreover, Uro B (50 μM) could inhibit the TLR4, Src, IκBα, NF-κBp65 and their phosphorylated protein expression (P < 0.05). Therefore, Uro B may inhibit macrophage M1 polarization via miR155-5p mediated MAPK/NF-кB pathway.
Collapse
Affiliation(s)
| | - Teng-Fei Ji
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wan-Ting Ding
- School of Pharmacy, Xinjiang Medical University, Urumqi 830046, China
| | - Chen-Yang Li
- Key Laboratory for Uighur Medicine, Institute of Materia Medica of Xinjiang, Urumqi 830004, China
| | | | - Ren Ci
- Hospital of Tibetan Traditional Medicine, Lhasa 850002, China
| | - Jun Zhao
- School of Pharmacy, Xinjiang Medical University, Urumqi 830046, China
- Key Laboratory for Uighur Medicine, Institute of Materia Medica of Xinjiang, Urumqi 830004, China
| |
Collapse
|
15
|
Huang Z, Zhang W, Shu Q, Guo XC, Zheng X, Lu YJ. Synergistic Anti-Inflammatory Effects of Dibenzoylmethane and Silibinin: Insights From LPS-Induced RAW 264.7 Cells and TPA-Induced Mouse Model. Chem Biodivers 2025; 22:e202402567. [PMID: 39743480 DOI: 10.1002/cbdv.202402567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/11/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Inflammation is an important predisposing factor for many chronic diseases. The dietary flavonoid silibinin (SB) has excellent anti-inflammatory properties in cells, but its low bioavailability in the blood compromises its therapeutic potential. This study aims to investigate the potential of dibenzoylmethane (DBM) to synergistically enhance the anti-inflammatory benefits of SB. The synergistic effects of DBM and SB in combination were evaluated in lipopolysaccharide (LPS)-induced RAW264.7 cells and 12-O-tetradecanoylphorbol 13-acetate (TPA)-induced mice. In addition, a network pharmacology approach and molecular docking were used to explore the key targets and signaling pathways of DBM and SB in combination. The results showed that DBM and SB synergistically inhibited the production of nitric oxide (NO), reactive oxygen species (ROS), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α) in a 1:1 concentration ratio. These two compounds may exert their synergistic effects by modulating the nuclear factor kappa-B (NF-κB) and HIF-1 signaling pathways, among others. Molecular docking revealed that both compounds exhibited high binding affinities to inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Compared with single-compound use, the two compounds in combination significantly reduced ear edema and inflammatory cell infiltration and inhibited the protein expression of iNOS and COX-2 in TPA-induced mice. This research provides a rationale for the combination of DBM and SB as an effective anti-inflammatory agent.
Collapse
Affiliation(s)
- Zebin Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Wanying Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Qi Shu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Xiao-Chun Guo
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Xi Zheng
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Yu-Jing Lu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
- Guangdong Xianlingtong Biopharmaceutical Technology Co., Ltd, Meizhou, China
| |
Collapse
|
16
|
Perner F, Pahl HL, Zeiser R, Heidel FH. Malignant JAK-signaling: at the interface of inflammation and malignant transformation. Leukemia 2025; 39:1011-1030. [PMID: 40140631 PMCID: PMC12055591 DOI: 10.1038/s41375-025-02569-8] [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: 01/02/2025] [Revised: 02/21/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025]
Abstract
The JAK pathway is central to mammalian cell communication, characterized by rapid responses, receptor versatility, and fine-tuned regulation. It involves Janus kinases (JAK1, JAK2, JAK3, TYK2), which are activated when natural ligands bind to receptors, leading to autophosphorylation and activation of STAT transcription factors [1, 2]. JAK-dependent signaling plays a pivotal role in coordinating cell communication networks across a broad spectrum of biological systems including development, immune responses, cell growth, and differentiation. JAKs are frequently mutated in the aging hematopoietic system [3, 4] and in hematopoietic cancers [5]. Thus, dysregulation of the pathway results in various diseases, including cancers and immune disorders. The binding of extracellular ligands to class I and II cytokine receptors initiates a critical signaling cascade through the activation of Janus kinases (JAKs). Upon ligand engagement, JAKs become activated and phosphorylate specific tyrosine residues on the receptor, creating docking sites for signal transducer and activator of transcription (STAT) proteins. Subsequent JAK-mediated phosphorylation of STATs enables their dimerization and nuclear translocation, where they function as transcription factors to modulate gene expression. Under physiological conditions, JAK-signaling is a tightly regulated mechanism that governs cellular responses to external cues, such as cytokines and growth factors, ensuring homeostasis and maintaining the functional integrity of tissues and organs. Highly defined regulation of JAK-signaling is essential for balancing cellular responses to inflammatory stimuli and growth signals, thus safeguarding tissue health. In contrast, dysregulated JAK-signaling results in chronic inflammation and unrestrained cellular proliferation associated with various diseases. Understanding the qualitative and quantitative differences at the interface of physiologic JAK-signaling and its aberrant activation in disease is crucial for the development of targeted therapies that precisely tune this pathway to target pathologic activation patterns while leaving homeostatic processes largely unaffected. Consequently, pharmaceutical research has targeted this pathway for drug development leading to the approval of several substances with different selectivity profiles towards individual JAKs. Yet, the precise impact of inhibitor selectivity and the complex interplay of different functional modules within normal and malignant cells remains incompletely understood. In this review, we summarize the current knowledge on JAK-signaling in health and disease and highlight recent advances and future directions in the field.
Collapse
Affiliation(s)
- Florian Perner
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany
| | - Heike L Pahl
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian H Heidel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany.
- Leibniz-Institute on Aging, Fritz-Lipmann-Institute (FLI), Jena, Germany.
- Cellular Therapy Center (CTC), Hannover Medical School (MHH), Hannover, Germany.
| |
Collapse
|
17
|
Chen YF, Lu YQ, Gao WY, Fan BY, Ren FC, Shen CP. Anti-inflammatory abietane-type diterpenoids from the roots of Salvia przewalskii. PHYTOCHEMISTRY 2025; 237:114523. [PMID: 40311889 DOI: 10.1016/j.phytochem.2025.114523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/03/2025]
Abstract
Fourteen diterpenes were isolated and purified from the roots of Salvia przewalskii, including eight previously unreported abietane analogues (compounds 1-4, 9, 10a, 11a, and 11b), five known ones (5-8 and 10b), and one icetexane analogue (12). The structures were determined through spectroscopic data interpretation, optical rotations, calculated NMR-DP4+ analysis, and ECD. Compounds 1-8 belong to a class of abietane derivatives containing a [5, 5]-oxospirolactone moiety. The research explored the mechanism behind the prevalence of 16β-type [5, 5]-oxospirolactone as the major component in the inseparable mixtures, shedding light on the proposed biosynthetic pathway of these compounds. Consistent with experimental findings, it was revealed that the 16β-type [5, 5]-oxospirolactone was shown to exhibit significantly lower free energy of formation compared to the 16α-product. Additionally, all compounds were evaluated for their ability to inhibit NO production in LPS-induced RAW 264.7 macrophages. Compounds 1, 2, and 11a demonstrated promising bioactive properties in terms of inhibiting NO release.
Collapse
Affiliation(s)
- Yan-Fang Chen
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Yu-Qing Lu
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Wan-Yun Gao
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Bo-Yi Fan
- School of Pharmacy, Nantong University, 9 Seyuan Road, Nantong, 226019, People's Republic of China
| | - Fu-Cai Ren
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China.
| | - Chuan-Pu Shen
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China.
| |
Collapse
|
18
|
Doroszkiewicz J, Winkel I, Mroczko B. Comparative analysis of neuroinflammatory pathways in Alzheimer's disease, Parkinson's disease, and multiple sclerosis: insights into similarities and distinctions. Front Neurosci 2025; 19:1579511. [PMID: 40364858 PMCID: PMC12069400 DOI: 10.3389/fnins.2025.1579511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Neurodegenerative diseases, contributing to the significant socioeconomic burden due to aging society, are gaining increasing interest. Despite each disease having different etiologies, neuroinflammation is believed to play a crucial role in Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). In addition to the pathogenic function of inflammation in the brain there is growing evidence that immune responses are essential for neuroregeneration. This review compares and contrasts the neuroinflammatory pathways that selected neurodegenerative diseases share and have in common. In AD, tau tangles and beta-amyloid plaques cause microglia and astrocytes to become activated in an inflammatory response. Alpha-synuclein aggregation stimulate neuroinflammation in Parkinson's disease, especially in the substantia nigra. In Multiple Sclerosis an autoimmune attack on myelin is connected to inflammation via invading immune cells. Commonalities include the release of pro-inflammatory mediators like cytokines and activation of signaling pathways such as NF-κB and MAPK. Comprehending these common routes is essential for discovering early diagnostic possibilities for the diseases and possible tailored treatments. Our work underscores the potential for insights into disease mechanisms. Identifying common targets offers promise for advancing our understanding and potential future treatment approaches across these debilitating disorders.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Izabela Winkel
- Dementia Disorders Centre, Medical University of Wroclaw, Scinawa, Poland
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
19
|
Rosell A, Krygowska AA, Alcón Pérez M, Cuesta C, Voisin MB, de Paz J, Sanz-Fraile H, Rajeeve V, Carreras-González A, Berral-González A, Swinyard O, Gabandé-Rodríguez E, Downward J, Alcaraz J, Anguita J, García-Macías C, De Las Rivas J, Cutillas PR, Castellano Sanchez E. RAS-p110α signalling in macrophages is required for effective inflammatory response and resolution of inflammation. eLife 2025; 13:RP94590. [PMID: 40272400 PMCID: PMC12021417 DOI: 10.7554/elife.94590] [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] [Indexed: 04/25/2025] Open
Abstract
Macrophages are crucial in the body's inflammatory response, with tightly regulated functions for optimal immune system performance. Our study reveals that the RAS-p110α signalling pathway, known for its involvement in various biological processes and tumourigenesis, regulates two vital aspects of the inflammatory response in macrophages: the initial monocyte movement and later-stage lysosomal function. Disrupting this pathway, either in a mouse model or through drug intervention, hampers the inflammatory response, leading to delayed resolution and the development of more severe acute inflammatory reactions in live models. This discovery uncovers a previously unknown role of the p110α isoform in immune regulation within macrophages, offering insight into the complex mechanisms governing their function during inflammation and opening new avenues for modulating inflammatory responses.
Collapse
Affiliation(s)
- Alejandro Rosell
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Agata Adelajda Krygowska
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Marta Alcón Pérez
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Cristina Cuesta
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Mathieu-Benoit Voisin
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Juan de Paz
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Héctor Sanz-Fraile
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de BarcelonaBarcelonaSpain
| | - Vinothini Rajeeve
- Centre for Cancer Genomics and Computational Biology, Cell Signalling and Proteomics Laboratory, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Ana Carreras-González
- Bioinformatics and Functional Genomics, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de SalamancaSalamancaSpain
| | | | - Ottilie Swinyard
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Enrique Gabandé-Rodríguez
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Julian Downward
- Oncogene Biology Laboratory, Francis Crick InstituteLondonUnited Kingdom
| | - Jordi Alcaraz
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de BarcelonaBarcelonaSpain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST)BarcelonaSpain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Lab, CIC bioGUNEDerioSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
- Pathology Unit, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de SalamancaSalamancaSpain
| | - Carmen García-Macías
- Pathology Unit, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de SalamancaSalamancaSpain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de SalamancaSalamancaSpain
| | - Pedro R Cutillas
- Centre for Cancer Genomics and Computational Biology, Cell Signalling and Proteomics Laboratory, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Esther Castellano Sanchez
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| |
Collapse
|
20
|
Tang Y, Guo T, Wang X, Li C, Zhang X, Zhang J. Cyclodextrin-Derived Macromolecular Therapies for Inflammatory Diseases. Macromol Biosci 2025:e2400637. [PMID: 40271896 DOI: 10.1002/mabi.202400637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Inflammation is an essential physiological defense mechanism against harmful stimuli, yet dysregulated inflammatory responses are closely associated with the pathogenesis of numerous acute and chronic diseases. Recent advances highlight the remarkable anti-inflammatory potential of bioactive macromolecules, particularly cyclodextrins (CDs) and their engineered derivatives, which are emerging as promising therapeutic agents. This review systematically introduces different CDs and CD-derived macromolecules that demonstrate anti-inflammatory properties, with emphasis on their molecular mechanisms of action. Native CDs exhibit direct therapeutic effects through host-guest interactions, enabling selective sequestration of pathogenic components such as cholesterol crystals and proteins that drive inflammatory cascades. Moreover, chemically modified CD derivatives incorporating functional groups demonstrate enhanced capabilities in neutralizing inflammatory mediators and modulating immune cell responses. This work further discusses the expanding therapeutic applications of these macromolecules across diverse inflammatory conditions, ranging from acute tissue injuries to chronic autoimmune disorders. Finally, this work critically analyzes the crucial challenges and emerging opportunities in translating CD-based macromolecular therapies into clinical practice, addressing key considerations in biocompatibility, targeted delivery, and therapeutic efficacy optimization.
Collapse
Affiliation(s)
- Yige Tang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- International Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Tao Guo
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xuanran Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiangjun Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Yu-Yue Pathology Scientific Research Center, 313 Gaoteng Avenue, Jiulongpo District, Chongqing, 400039, China
| |
Collapse
|
21
|
Manoharan MS, Lee GC, Harper N, Meunier JA, Restrepo MI, Jimenez F, Karekatt S, Branum AP, Gaitan AA, Andampour K, Smith AM, Mader M, Noronha M, Tripathy D, Zhang N, Moreira AG, Pandranki L, Sanchez-Reilly S, Trinh HD, Barnett C, Angel L, Segal LN, Nicholson S, Clark RA, He W, Okulicz JF, Ahuja SK. The 15-Year Survival Advantage: Immune Resilience as a Salutogenic Force in Healthy Aging. Aging Cell 2025:e70063. [PMID: 40264357 DOI: 10.1111/acel.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Human aging presents an evolutionary paradox: while aging rates remain constant, healthspan and lifespan vary widely. We address this conundrum via salutogenesis-the active production of health-through immune resilience (IR), the capacity to resist disease despite aging and inflammation. Analyzing ~17,500 individuals across lifespan stages and inflammatory challenges, we identified a core salutogenic mechanism: IR centered on TCF7, a conserved transcription factor maintaining T-cell stemness and regenerative potential. IR integrates innate and adaptive immunity to counter three aging and mortality drivers: chronic inflammation (inflammaging), immune aging, and cellular senescence. By mitigating these aging mechanisms, IR confers survival advantages: At age 40, individuals with poor IR face a 9.7-fold higher mortality rate-a risk equivalent to that of 55.5-year-olds with optimal IR-resulting in a 15.5-year gap in survival. Optimal IR preserves youthful immune profiles at any age, enhances vaccine responses, and reduces burdens of cardiovascular disease, Alzheimer's, and serious infections. Two key salutogenic evolutionary themes emerge: first, female-predominant IR, including TCF7, likely reflects evolutionary pressures favoring reproductive success and caregiving; second, midlife (40-70 years) is a critical window where optimal IR reduces mortality by 69%. After age 70, mortality rates converge between resilient and non-resilient groups, reflecting biological limits on longevity extension. TNFα-blockers restore salutogenesis pathways, indicating IR delays aging-related processes rather than altering aging rates. By reframing aging as a salutogenic-pathogenic balance, we establish TCF7-centered IR as central to healthy longevity. Targeted midlife interventions to enhance IR offer actionable strategies to maximize healthspan before biological constraints limit benefits.
Collapse
Affiliation(s)
- Muthu Saravanan Manoharan
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Grace C Lee
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Pharmacotherapy Education and Research Center, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Nathan Harper
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Justin A Meunier
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Marcos I Restrepo
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Fabio Jimenez
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Sreenath Karekatt
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Anne P Branum
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Alvaro A Gaitan
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | - Kian Andampour
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Alisha M Smith
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Michael Mader
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Michelle Noronha
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Devjit Tripathy
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Nu Zhang
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Alvaro G Moreira
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Lavanya Pandranki
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Sandra Sanchez-Reilly
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Hanh D Trinh
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Clea Barnett
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Luis Angel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Leopoldo N Segal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Susannah Nicholson
- Department of Surgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Robert A Clark
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Weijing He
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- The Foundation for Advancing Veterans' Health Research, San Antonio, Texas, USA
| | | | - Sunil K Ahuja
- Veterans Affairs Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, San Antonio, Texas, USA
| |
Collapse
|
22
|
Ma XC, Clardy J. Spontaneous Generation of an Endogenous RORγt Agonist. J Am Chem Soc 2025; 147:11688-11692. [PMID: 40145418 PMCID: PMC11987011 DOI: 10.1021/jacs.5c02724] [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: 02/13/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 03/28/2025]
Abstract
The transcription factor RORγt regulates the development of Th17 cells and their inflammatory cytokine IL-17─a pathway that can both clear bacterial pathogens and drive autoimmune diseases. An endogenous RORγt agonist with a noncanonical structure, a lysophosphatidylethanolamine (1-18:1-LPE or 1), was recently identified, and its identity both increases our understanding of immune regulation and creates options for therapeutic intervention. Compound 1 could be formed directly through enzymatic cleavage of a suitable phosphatidylethanolamine (PE) by a phospholipase A2 (PLA2) or by "triggering" of a suitable plasmalogen with accompanying 1,2-acyl migration from the sn-2 to sn-1 positions of glycerol. This study illustrates the plausibility of a plasmalogen-based pathway through synthesis of the plasmalogen precursor (2) and triggering the plasmalogen's electron-rich vinyl ether with small electrophiles characteristic of inflammatory and tumor environments to create 1-18:1-LPE (1). The plasmalogen-based pathway is consistent with previous studies on the formation of 1, and it also conforms to Lands rules for acyl chain distribution and provides a mechanism for immune signaling with both spatial and temporal control.
Collapse
Affiliation(s)
- Xiao Corey Ma
- Department
of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, United States
- Laboratory
of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jon Clardy
- Department
of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
23
|
Bourgeois JS, McCarthy JE, Turk SP, You SS, Bernard Q, Clendenen LH, Wormser GP, Marcos LA, Dardick K, Telford SR, Marques AR, Hu LT. Peromyscus leucopus, Mus musculus, and humans have distinct transcriptomic responses to larval Ixodes scapularis bites. Infect Immun 2025; 93:e0006525. [PMID: 40066985 PMCID: PMC11977304 DOI: 10.1128/iai.00065-25] [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: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Ixodes scapularis ticks are an important vector for at least seven tick-borne human pathogens, including a North American Lyme disease spirochete, Borrelia burgdorferi. The ability for these ticks to survive in nature is credited, in part, to their ability to feed on a variety of hosts without triggering an immune response capable of preventing tick feeding. While the ability of nymphal ticks to feed on a variety of hosts has been well documented, the host-parasite interactions between larval I. scapularis and different vertebrate hosts are relatively unexplored. Here we report on the changes in the vertebrate host transcriptome present at the larval tick bite site using the natural I. scapularis host Peromyscus leucopus, a non-natural rodent host, Mus musculus (BALB/c), and humans. We note substantially less evidence of activation of canonical proinflammatory pathways in P. leucopus compared to BALB/c mice and pronounced evidence of inflammation in humans. Pathway enrichment analyses revealed a particularly strong signature of interferon gamma, tumor necrosis factor, and interleukin 1 signaling at the BALB/c and human tick bite sites. We also note that bite sites on BALB/c mice and humans, but not deer mice, show activation of wound-healing pathways. These data provide molecular evidence of the coevolution between larval I. scapularis and P. leucopus and, in addition, expand our overall understanding of I. scapularis feeding.
Collapse
Affiliation(s)
- Jeffrey S. Bourgeois
- Tufts Lyme Disease Initiative, Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Julie E. McCarthy
- Tufts Lyme Disease Initiative, Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Siu-Ping Turk
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Stephanie S. You
- Tufts Lyme Disease Initiative, Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Quentin Bernard
- Tufts Lyme Disease Initiative, Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Luke H. Clendenen
- Tufts Lyme Disease Initiative, Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Gary P. Wormser
- Division of Infectious Diseases, Department of Medicine, New York Medical College, Valhalla, New York, USA
| | - Luis A. Marcos
- Department of Medicine, Division of Infectious Diseases, Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | | | - Sam R. Telford
- Tufts Lyme Disease Initiative, Department of Infectious Disease and Global Health, Tufts University, North Grafton, Massachusetts, USA
| | - Adriana R. Marques
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Linden T. Hu
- Tufts Lyme Disease Initiative, Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Carra D, Maas SCE, Seoane JA, Alonso-Curbelo D. Exposomal determinants of non-genetic plasticity in tumor initiation. Trends Cancer 2025; 11:295-308. [PMID: 40023688 DOI: 10.1016/j.trecan.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 03/04/2025]
Abstract
The classical view of cancer as a genetically driven disease has been challenged by recent findings of oncogenic mutations in phenotypically healthy tissues, refocusing attention on non-genetic mechanisms of tumor initiation. In this context, gene-environment interactions take the stage, with recent studies showing how they unleash and redirect cellular and tissue plasticity towards protumorigenic states in response to the exposome, the ensemble of environmental factors impinging on tissue homeostasis. We conceptualize tumor-initiating plasticity as a phenotype-transforming force acting at three levels: cell-intrinsic, focusing on mutant epithelial cells' responses to environmental variation; reprogramming of non-neoplastic cells of the host, leading to protumor micro- and macroenvironments; and microbiome ecosystem dynamics. This perspective highlights cell, tissue, and organismal plasticity mechanisms underlying tumor initiation that are shaped by the exposome, and how their functional investigation may provide new opportunities to prevent, detect, and intercept cancer-promoting plasticity.
Collapse
Affiliation(s)
- Davide Carra
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Silvana C E Maas
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jose A Seoane
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Direna Alonso-Curbelo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| |
Collapse
|
25
|
Juaim AN, Sun J, Nie R, Li W, Ding L, Wang K, Zhou J, Li M, Chi M, Dong B, Qi M, Wang L. IR820 Sensitized Ceria Nanozyme via PDA Bridging for Multifaceted Antibacterial Wound Healing Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2500382. [PMID: 40159834 DOI: 10.1002/smll.202500382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/20/2025] [Indexed: 04/02/2025]
Abstract
Nanozymes with peroxidase (POD)-like activity hold significant potential for addressing antibiotic-resistant bacterial infections. However, their catalytic efficiency and therapeutic efficacy need further improvement to broaden their clinical applications. A key challenge is achieving efficient energy transfer from photosensitizing molecules to nanozymes, which is critical for enhancing catalytic performance. In this study, a universal strategy is developed to bridge nanozymes and photosensitizing molecules, designing photoactivated nanozymes called IR820/PDA@mCeO2 (IR/P@Ce). By integrating IR820, a photosensitizer, with mesoporous ceria (mCeO2), it facilitates efficient electron transfer through polydopamine (PDA) bridge molecules, resulting in enhanced POD-like catalytic performance and reactive oxygen species production. Additionally, PDA stabilized the nanozyme, improved photothermal therapy, and enhanced photodynamic therapy under near-infrared light exposure, further amplifying bacterial destruction. This multifunctional nanozyme demonstrated strong antibacterial efficacy against both Gram-positive (Staphylococcus aureus) and Gram-negative (Escherichia coli) bacteria. Moreover, its synergistic approach not only facilitated bacterial eradication but also accelerated wound healing in vivo, making it a promising therapeutic alternative for managing bacterial infections and promoting tissue regeneration.
Collapse
Affiliation(s)
- Akram Nasser Juaim
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Department of Prosthodontics, College of Dentistry, Thamar University, Dhamar, 87246, Yemen
| | - Jiao Sun
- Department of Cell Biology, Norman Bethune College of Medicine, Jilin University, Changchun, 130021, China
| | - Ran Nie
- Department of Stomatology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Wen Li
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Lina Ding
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Kun Wang
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Jing Zhou
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Meiqi Li
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Minghan Chi
- School of Dentistry, University of Minnesota, Malcom Moos Health Sciences Tower, 515 Delaware St SE, Minneapolis, MN, 55455, USA
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Manlin Qi
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Lin Wang
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Sciences and Technology for Stomatology Nanoengineering, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| |
Collapse
|
26
|
Russo RC, Togbe D, Couillin I, Segueni N, Han L, Quesniaux VFJ, Stoeger T, Ryffel B. Ozone-induced lung injury and inflammation: Pathways and therapeutic targets for pulmonary diseases caused by air pollutants. ENVIRONMENT INTERNATIONAL 2025; 198:109391. [PMID: 40121788 DOI: 10.1016/j.envint.2025.109391] [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: 08/23/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Exposure to ambient Ozone (O3) air pollution directly causes by its oxidative properties, respiratory epithelial cell injury, and cell death, which promote inflammation and hyperreactivity, posing a significant public health concern. Recent clinical and experimental studies have made strides in elucidating the mechanisms underlying O3-induced epithelial cell injury, inflammation, and airway hyperreactivity, which are discussed herein. The current data suggest that O3-induced oxidative stress is a central event-inducing oxeiptotic cell death pathway. O3-induced epithelial barrier damage and cell death, triggering the release of alarmins and damage-associated molecular patterns (DAMPs), with subsequent endogenous activation of Toll-like receptors (TLRs), DNA sensing pathways, and inflammasomes, activating interleukin-1-Myd88 inflammatory pathway with the production of a range of chemokines and cytokines. This cascade orchestrates lung tissue-resident cell activation in response to O3 in leukocyte and non-leukocyte populations, driving sterile innate immune response. Chronic inflammatory response to O3, by repeated exposures, supports a mixed phenotype combining asthma and emphysema, in which their exacerbation by other particulate pollutants potentially culminates in respiratory failure. We use data from lung single-cell transcriptomics to map genes of O3-damage sensing and signaling pathways to lung cells and thereby highlight potential hotspots of O3 responses. Deeper insights into these pathological pathways might be helpful for the identification of novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Remo C Russo
- Laboratory of Pulmonary Immunology and Mechanics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Dieudonnée Togbe
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France
| | - Isabelle Couillin
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France
| | | | - Lianyong Han
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center, Helmholtz Zentrum München, German Research Center for Environmental Health, and Member of the German Center of Lung Research (DZL), Germany
| | - Valérie F J Quesniaux
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France
| | - Tobias Stoeger
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center, Helmholtz Zentrum München, German Research Center for Environmental Health, and Member of the German Center of Lung Research (DZL), Germany
| | - Bernhard Ryffel
- Laboratory of Immuno-Neuro Modulation, INEM, UMR7355 CNRS and University of Orleans, Orleans, France; ArtImmune SAS, 13 Avenue Buffon, Orleans, France.
| |
Collapse
|
27
|
Han Z, Fu J, Gong A, Ren W. Bacterial indole-3-propionic acid inhibits macrophage IL-1β production through targeting methionine metabolism. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1118-1131. [PMID: 39825207 DOI: 10.1007/s11427-024-2789-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/20/2024] [Indexed: 01/20/2025]
Abstract
The gut microbiota plays key roles in host health by shaping the host immune responses through their metabolites, like indole derivatives from tryptophan. However, the direct role of these indole derivatives in macrophage fate decision and the underlying mechanism remains unknown. Here, we found that bacterial indole-3-propionic acid (IPA) downregulates interleukin-1beta (IL-1β) production in M1 macrophages through inhibition of nuclear factor-kappa B (NF-κB) signaling. Mechanistically, IPA binds specifically with methionine adenosyl-transferase 2A (MAT2A) to promote S-adenosylmethionine (SAM) synthesis, which facilitates the DNA methylation of ubiquitin-specific peptidase 16 (USP16, a deubiquitinase), and in turn promotes Toll-like receptor 4 (TLR4) ubiquitination and NF-κB inhibition. Furthermore, IPA administration attenuates sepsis in mouse models induced by lipopolysaccharides (LPS), showcasing its potential as a microbial-derived adjunct in alleviating inflammation. Collectively, our findings reveal a newly found microbial metabolite-immune system regulatory pathway mediated by IPA.
Collapse
Affiliation(s)
- Ziyi Han
- State Key Laboratory of Livestock and Poultry Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Animal Disease-resistant Nutrition, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 625014, China
| | - Jian Fu
- State Key Laboratory of Livestock and Poultry Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Aiyan Gong
- China Institute of Veterinary Drug Control, Beijing, 100081, China
| | - Wenkai Ren
- State Key Laboratory of Livestock and Poultry Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
28
|
Kuo PJ, Chen LT, Li SM, Chen ZR, Chen JJ. Pharmacological and Molecular Docking Investigation of Leaves of Eriobotrya japonica: Antioxidant, Enzyme Inhibition, and Anti-Inflammatory Effects. Antioxidants (Basel) 2025; 14:413. [PMID: 40298661 PMCID: PMC12024191 DOI: 10.3390/antiox14040413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Leaves of Eriobotrya japonica have long been utilized in traditional Chinese medicine (TCM) for treating pulmonary inflammation and stomach disorders. This study extends their pharmacological applications by evaluating the antioxidant, anti-α-glucosidase, anti-acetylcholinesterase (AChE), and anti-inflammatory activities of solvent extracts and isolated bioactive components through an integrative approach combining extraction, bioassays, and molecular docking. Solvent extracts prepared with varying polarities exhibited distinct bioactivities, with the 100 °C water and methanol extracts displaying the strongest antioxidant potential. The ethyl acetate extract exhibited potent α-glucosidase inhibition, whereas the n-hexane extract demonstrated significant AChE inhibitory activity. Among the isolated compounds, epicatechin (5) (SC50 = 7.83 ± 0.34 μM) and rutin (6) (SC50 = 6.69 ± 0.25 μM) showed superior ABTS and superoxide scavenging activities, respectively, compared to the positive controls (BHT and cynaroside). Ursolic acid (2) exhibited stronger α-glucosidase inhibition (IC50 = 10.68 ± 0.76 μM) than acarbose (IC50 = 419.93 ± 29.15 μM), while tormentic acid (4) demonstrated superior AChE inhibition compared to chlorogenic acid. Ursolic acid (2) also displayed NO inhibition (IC50 = 20.18 ± 1.46 μM) comparable to quercetin (IC50 = 17.05 ± 1.63 μM), with Western blot analysis confirming its potent iNOS inhibitory activity. Molecular docking further supported these findings, revealing that ursolic acid (2) exhibited stronger binding affinity to α-glucosidase (-8.7 kcal/mol) than acarbose (-5.1 kcal/mol), tormentic acid (4) displayed higher binding energy to AChE (-8.8 kcal/mol) compared to chlorogenic acid (-7.8 kcal/mol), and ursolic acid (2) (-7.5 kcal/mol) showed a binding affinity to iNOS similar to that of quercetin (-7.7 kcal/mol). These results highlight the strong potential of E. japonica leaf extracts and bioactive compounds as natural antioxidants, enzyme inhibitors, and anti-inflammatory agents, supporting their development as dietary supplements or therapeutic candidates for managing oxidative stress, hyperglycemia, neurodegenerative diseases, and inflammatory disorders.
Collapse
Affiliation(s)
- Pao-Jen Kuo
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Li-Ting Chen
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-T.C.); (S.-M.L.); (Z.-R.C.)
| | - Sin-Min Li
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-T.C.); (S.-M.L.); (Z.-R.C.)
| | - Zih-Rong Chen
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-T.C.); (S.-M.L.); (Z.-R.C.)
| | - Jih-Jung Chen
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-T.C.); (S.-M.L.); (Z.-R.C.)
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404333, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
| |
Collapse
|
29
|
Hong H, Wu Y, Li Y, Han Y, Cao X, Wu VWY, Chan TTH, Zhou J, Cao Q, Lui KO, Wong CK, Dai Z, Tian XY. Endothelial PPARδ Ablation Exacerbates Vascular Hyperpermeability via STAT1/CXCL10 Signaling in Acute Lung Injury. Circ Res 2025; 136:735-751. [PMID: 39996324 DOI: 10.1161/circresaha.124.325855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/16/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND Vascular hyperpermeability is one of the hallmarks of acute lung injury, contributing to excessive inflammation and respiratory failure. The PPARδ (peroxisome proliferator-activated receptor delta) is an anti-inflammatory transcription factor, although its role in endothelial barrier function remains unclear. Here, we studied the essential role of PPARδ in maintaining vascular endothelial barrier integrity during lung inflammation and investigated the underlying mechanisms. METHODS Endothelial cell (EC)-selective PPARδ knockout mice (PpardEC-KO) and littermate control mice (PpardEC-WT) received lipopolysaccharide injection to induce acute lung injury. Lung inflammation, pulmonary vascular leakage, and mouse mortality were monitored. Single-cell RNA sequencing was performed on sorted mouse lung ECs. RESULTS PpardEC-KO mice exhibited aggravated lung inflammation, characterized by increased leukocyte infiltration, elevated production of proinflammatory cytokines, and higher mortality rates. The enhanced inflammatory responses were associated with increased protein leakage, interstitial edema, and impaired endothelial barrier structure, leading to vascular hyperpermeability in PpardEC-KO mice. Mechanistically, with single-cell RNA sequencing, we identified the emergence of an interferon-activated capillary EC population marked by CXCL10 (C-X-C motif chemokine 10) expression following lipopolysaccharide challenge. PPARδ silencing significantly increased CXCL10 expression in ECs through activating STAT1 (Signal transducer and activator of transcription 1). Notably, CXCL10 treatment induced degradation of tight junction proteins ZO-1 (zonula occludens protein 1) and claudin-5 through the ubiquitin-proteasome system, disrupting membrane junction continuity in ECs. Administration of anti-CXCL10 antibody or CXCL10 receptor antagonist AMG487 suppressed both lipopolysaccharide-induced lung inflammation and vascular leakage in PpardEC-KO mice. CONCLUSIONS These results highlighted a novel anti-inflammatory role of PPARδ in ECs by suppressing CXCL10-mediating vascular hyperpermeability. Targeting the CXCL10 signaling shows therapeutic potential against vascular injury in acute lung injury.
Collapse
Affiliation(s)
- Huiling Hong
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Yalan Wu
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, China (Y.W.)
| | - Yangxian Li
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Yumeng Han
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Xiaoyun Cao
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
- Department of Chemical Pathology (X.C., K.O.L., C.-K.W.), The Chinese University of Hong Kong
| | - Vivian Wei Yan Wu
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Thomas Ting Hei Chan
- School of Biomedical Sciences (T.T.H.C., J.Z., Q.C.), The Chinese University of Hong Kong
| | - Jingying Zhou
- School of Biomedical Sciences (T.T.H.C., J.Z., Q.C.), The Chinese University of Hong Kong
| | - Qin Cao
- School of Biomedical Sciences (T.T.H.C., J.Z., Q.C.), The Chinese University of Hong Kong
| | - Kathy O Lui
- Department of Chemical Pathology (X.C., K.O.L., C.-K.W.), The Chinese University of Hong Kong
| | - Chun-Kwok Wong
- Department of Chemical Pathology (X.C., K.O.L., C.-K.W.), The Chinese University of Hong Kong
| | - Zhiyu Dai
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona (Z.D.)
| | - Xiao Yu Tian
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| |
Collapse
|
30
|
García-Domínguez M. The Role of IL-23 in the Development of Inflammatory Diseases. BIOLOGY 2025; 14:347. [PMID: 40282212 PMCID: PMC12025033 DOI: 10.3390/biology14040347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Interleukin-23 is crucial in the initiation and progression of certain inflammatory disorders. As a key cytokine, IL-23 is involved in the differentiation and activation of Th17 cells, which play a role in a broad spectrum of inflammatory diseases. This review examines the molecular mechanisms through which IL-23 contributes to the pathogenesis of conditions including psoriasis, rheumatoid arthritis, inflammatory bowel disease, and multiple sclerosis. By elucidating the significant role of IL-23 in inflammation, this review underscores its importance as a therapeutic target for managing inflammatory conditions, with particular emphasis on current and emerging biologic treatments.
Collapse
Affiliation(s)
- Mario García-Domínguez
- Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain;
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
31
|
Metwally H. STAT Signature Dish: Serving Immunity with a Side of Dietary Control. Biomolecules 2025; 15:487. [PMID: 40305224 PMCID: PMC12024614 DOI: 10.3390/biom15040487] [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: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Immunity is a fundamental aspect of animal biology, defined as the host's ability to detect and defend against harmful pathogens and toxic substances to preserve homeostasis. However, immune defenses are metabolically demanding, requiring the efficient allocation of limited resources to balance immune function with other physiological and developmental needs. To achieve this balance, organisms have evolved sophisticated signaling networks that enable precise, context-specific responses to internal and external cues. These networks are essential for survival and adaptation in multicellular systems. Central to this regulatory architecture is the STAT (signal transducer and activator of Transcription) family, a group of versatile signaling molecules that govern a wide array of biological processes across eukaryotes. STAT signaling demonstrates remarkable plasticity, from orchestrating host defense mechanisms to regulating dietary metabolism. Despite its critical role, the cell-specific and context-dependent nuances of STAT signaling remain incompletely understood, highlighting a significant gap in our understanding. This review delves into emerging perspectives on immunity, presenting dynamic frameworks to explore the complexity and adaptability of STAT signaling and the underlying logic driving cellular decision-making. It emphasizes how STAT pathways integrate diverse physiological processes, from immune responses to dietary regulation, ultimately supporting organismal balance and homeostasis.
Collapse
Affiliation(s)
- Hozaifa Metwally
- Laboratory of Immune Regulation, The World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
32
|
Zhang T, Wang Y, Nie X, Chen X, Jin Y, Sun L, Yang R, Wang J, Xu W, Song T, Xie W, Chen X, Li C, Zhou J, Wu S, Li Y, Li T. ENKD1 modulates innate immune responses through enhanced geranylgeranyl pyrophosphate synthase activity. Cell Rep 2025; 44:115397. [PMID: 40048432 DOI: 10.1016/j.celrep.2025.115397] [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: 08/27/2024] [Revised: 12/22/2024] [Accepted: 02/14/2025] [Indexed: 03/29/2025] Open
Abstract
Inflammation is a crucial element of immune responses, with pivotal roles in host defenses against pathogens. Comprehensive understanding of the molecular mechanisms underlying inflammation is imperative for developing effective strategies to combat infectious diseases. Here, we conducted a screening analysis and identified enkurin domain-containing protein 1 (ENKD1) as a promising regulator of inflammation. We observed that ENKD1 expression was significantly reduced on activation of multiple Toll-like receptor (TLR) molecules. Deletion of ENKD1 resulted in enhanced innate immune system activation and exacerbation of septic inflammation. Mechanistically, ENKD1 interacted with geranylgeranyl diphosphate synthase 1 (GGPS1) and modulated its enzymatic activity, thereby influencing geranylgeranyl diphosphate production. This interaction ultimately led to Ras-related C3 botulinum toxin substrate 1 (RAC1) inactivation and suppression of pro-inflammatory signaling pathways. Our findings establish ENKD1 as a critical regulator of innate immune cell activation, underscoring its significant role in septic inflammation.
Collapse
Affiliation(s)
- Tianyu Zhang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Yixuan Wang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Xiaotong Nie
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Xiangrong Chen
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
| | - Yueyi Jin
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Lulu Sun
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Ruqian Yang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Jie Wang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Wenqing Xu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Ting Song
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Wei Xie
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiangfeng Chen
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
| | - Chaojun Li
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China; State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Sijin Wu
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China.
| | - Yan Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China.
| | - Tianliang Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China.
| |
Collapse
|
33
|
Zhang Q, Shen Y, Zhang C, Zhang H, Li X, Yang S, Dai C, Yu X, Lou J, Feng J, Hu C, Lin Z, Li X, Zhou X. Immunoengineered mitochondria for efficient therapy of acute organ injuries via modulation of inflammation and cell repair. SCIENCE ADVANCES 2025; 11:eadj1896. [PMID: 40106554 DOI: 10.1126/sciadv.adj1896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025]
Abstract
Acute organ injuries represent a major public health concern, driven by inflammation and mitochondrial dysfunction, leading to cell damage and organ failure. In this study, we engineered neutrophil membrane-fused mitochondria (nMITO), which combine the injury-targeting and anti-inflammatory properties of neutrophil membrane proteins with the cell repairing function of mitochondria. nMITO effectively blocked inflammatory cascades and restored mitochondrial function, targeting both key mechanisms in acute organ injuries. In addition, nMITO selectively targeted damaged endothelial cells via β-integrins and were delivered to injured tissues through tunneling nanotubes, enhancing their regulatory effects on inflammation and cell damage. In mouse models of acute myocardial injury, liver injury, and pancreatitis, nMITO notably reduced inflammatory responses and repaired tissue damage. These findings suggest that nMITO is a promising therapeutic strategy for managing acute organ injuries.
Collapse
Affiliation(s)
- Qing Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & Rehabilitation School, Kunming Medical University, Kunming 650500, PR China
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, PR China
| | - Yan Shen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Chengyuan Zhang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & Rehabilitation School, Kunming Medical University, Kunming 650500, PR China
| | - Hanyi Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Xuemei Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Shengqian Yang
- Institute of Materia Medica College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Chen Dai
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Xiuyan Yu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Jie Lou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Jinwei Feng
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Chenglu Hu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Zhihua Lin
- Chongqing University of Chinese Medicine, Chongqing 402760, PR China
| | - Xiaohui Li
- Institute of Materia Medica College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & Rehabilitation School, Kunming Medical University, Kunming 650500, PR China
- School of Pharmaceutical Sciences & Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, PR China
| |
Collapse
|
34
|
Wang Z, Li D, Wang Y, Yuan P, Zhang W, Zhang Y, He F, Yang J, Bi H, Duan H. Hyaluronic acid methacryloyl hydrogel with sustained IL-10 release promotes macrophage M2 polarization and motor function after spinal cord injury. J Biomater Appl 2025:8853282251329302. [PMID: 40111115 DOI: 10.1177/08853282251329302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
(1)Background: Inflammation plays a key role in spinal cord injury (SCI), where excessive inflammatory responses exacerbate neural damage and hinder regeneration. Modulating macrophage polarization, particularly through the sustained release of IL-10 to promote the anti-inflammatory M2 phenotype, represents a promising strategy to mitigate inflammation. In this study we developed a Hyaluronic Acid Methacryloyl (HAMA) hydrogel capable of sustained IL-10 release to regulate macrophage polarization and explore its therapeutic potential. (2)Methods: A photo-curable HAMA hydrogel was synthesized via methacrylation and designed for the sustained release of IL-10. The structural and functional properties were characterized using NMR and FT-IR. In vitro assays, including immunofluorescence, flow cytometry, and Western blotting, were performed to evaluate IL-10's effect on macrophage polarization. The anti-inflammatory and reparative effects of the hydrogel were further validated in a rat SCI. (3)Results: The HAMA hydrogel with sustained IL-10 release demonstrated excellent biocompatibility. It significantly promoted macrophage polarization to the anti-inflammatory M2 phenotype by increasing the expression of CD206. In vivo studies demonstrated that the group treated by HAMA with IL-10 exhibited recovery of sensory and motor functions, along with improvement of the inflammatory microenvironment at the site of injury. (4)Conclusion: The HAMA hydrogel with sustained IL-10 release effectively alleviates inflammation, enhances motor function after SCI, and serves as a promising immunomodulatory platform. This novel approach presents considerable potential for improving neural regeneration.
Collapse
Affiliation(s)
- Zhihua Wang
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Denghui Li
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yanghao Wang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ping Yuan
- Department of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wan Zhang
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yihe Zhang
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fei He
- Department of Orthopedics, Guandu District People's Hospital, Kunming, China
| | - Jianyi Yang
- Department of Orthopedics, Guandu District People's Hospital, Kunming, China
| | - Hangchuan Bi
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hao Duan
- Department of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
35
|
Hassan MAM, Fahmy MI, Azzam HN, Ebrahim YM, El-Shiekh RA, Aboulmagd YM. Multifaceted therapeutic potentials of catalpol, an iridoid glycoside: an updated comprehensive review. Inflammopharmacology 2025:10.1007/s10787-025-01694-1. [PMID: 40097877 DOI: 10.1007/s10787-025-01694-1] [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: 12/04/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Catalpol, classified as an iridoid glucoside, is recognized for its significant role in medicine, particularly in the treatment of various conditions such as diabetes mellitus, neuronal disorders, and inflammatory diseases. This review aims to evaluate the biological implications of catalpol and the mechanisms underlying its diverse pharmacological effects. A thorough exploration of existing literature was conducted utilizing the keyword "Catalpol" across prominent public domains like Google Scholar, PubMed, and EKB. Catalpol has demonstrated a diverse array of pharmacological effects in experimental models, showcasing its anti-diabetic, cardiovascular-protective, neuroprotective, anticancer, hepatoprotective, anti-inflammatory, and antioxidant properties. In summary, catalpol manifests a spectrum of biological effects through a myriad of mechanisms, prominently featuring its anti-inflammatory and antioxidant capabilities. Its diverse pharmacological profile underscores its potential for therapeutic applications across a range of conditions. Further research is warranted to fully elucidate the clinical implications of catalpol and optimize its use in medical practice.
Collapse
Affiliation(s)
- Mennat-Allah M Hassan
- Department of Pharmacology & Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Mohamed I Fahmy
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Hany N Azzam
- Pharmacy Practice Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Yasmina M Ebrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Yara M Aboulmagd
- Department of Pharmacology & Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
36
|
Emerson MJ, Willacy O, Madsen CD, Reuten R, Brøchner CB, Lund TK, Dahl AB, Jensen THL, Erler JT, Mayorca-Guiliani AE. Machine learning identifies remodeling patterns in human lung extracellular matrix. Acta Biomater 2025; 195:94-103. [PMID: 39746529 DOI: 10.1016/j.actbio.2024.12.062] [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: 08/08/2023] [Revised: 12/15/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Organ function depends on the three-dimensional integrity of the extracellular matrix (ECM). The structure resulting from the location and association of ECM components is a central regulator of cell behavior, but a dearth of matrix-specific analysis keeps it unresolved. Here, we deploy a high-resolution, 3D ECM mapping method and design a machine-learning powered pipeline to detect and characterize ECM architecture during health and disease. We deploy these tools in the human lung, an organ heavily dependent on ECM structure that can host diseases with different histopathologies. We analyzed segments from healthy, emphysema, usual interstitial pneumonia, sarcoidosis, and COVID-19 patients, and produced a remodeling signature per disease and a health/disease probability map from which we inferred the architecture of healthy and diseased ECM. Our methods demonstrate that exaggerated matrix deposition, or fibrosis, is not a single phenomenon, but a series of disease-specific alterations. STATEMENT OF SIGNIFICANCE: The extracellular matrix, or ECM, is the foremost biomaterial. It shapes and supports all tissues while regulating all cells. ECM structure is intricate, yet precise: each organ, at every stage, has a specific ECM structure. During disease, tissues suffer from structural changes that accelerate and perpetuate illness by dysregulating cells. Both healthy and diseased ECM structures are of great biomedical importance, but surprisingly, they have not been mapped in detail. Here, we present a method that combines tissue engineering with machine learning to reveal, map and analyze ECM structures, applied it to pulmonary diseases that kill millions every year. This method can bring objectivity and a higher degree of confidence into the diagnosis of pulmonary disease. In addition the amount of tissue needed for a firm diagnosis may be much smaller than required for manual microscopy evaluation.
Collapse
Affiliation(s)
- Monica J Emerson
- Section for Visual Computing, Department of Applied Mathematics and Computer Science, Technical University of Denmark. Kongens Lyngby, Denmark; Currently at Digital Science and Innovation, Novo Nordisk A/S, Måløv, Denmark
| | - Oliver Willacy
- Biotech Research and Innovation Center, Faculty of Health Sciences, University of Copenhagen. Copenhagen, Denmark; Department of Pathology, Rigshospitalet, University Hospital of Copenhagen. Copenhagen, Denmark
| | - Chris D Madsen
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University. Lund, Sweden; Currently at Symphogen A/S, Ballerup, Denmark
| | - Raphael Reuten
- Biotech Research and Innovation Center, Faculty of Health Sciences, University of Copenhagen. Copenhagen, Denmark; Currently at Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg. Freiburg, Germany
| | - Christian B Brøchner
- Department of Pathology, Rigshospitalet, University Hospital of Copenhagen. Copenhagen, Denmark
| | - Thomas K Lund
- Section for Lung Transplantation, Department of Cardiology, Rigshospitalet, University Hospital of Copenhagen. Copenhagen, Denmark
| | - Anders B Dahl
- Section for Visual Computing, Department of Applied Mathematics and Computer Science, Technical University of Denmark. Kongens Lyngby, Denmark
| | - Thomas H L Jensen
- Department of Pathology, Rigshospitalet, University Hospital of Copenhagen. Copenhagen, Denmark.
| | - Janine T Erler
- Biotech Research and Innovation Center, Faculty of Health Sciences, University of Copenhagen. Copenhagen, Denmark.
| | - Alejandro E Mayorca-Guiliani
- Biotech Research and Innovation Center, Faculty of Health Sciences, University of Copenhagen. Copenhagen, Denmark; Currently at Nordic Bioscience A/S. Herlev, Denmark.
| |
Collapse
|
37
|
Ji H, Kuang G, Yang H, Liu H, Li Y, Hu S, Xiao A, You C, Sun H, Fan C, Sun G. Discrepancies between human and murine model cerebral aneurysms at single-cell resolution. Front Cell Dev Biol 2025; 13:1512938. [PMID: 40134579 PMCID: PMC11933115 DOI: 10.3389/fcell.2025.1512938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/14/2025] [Indexed: 03/27/2025] Open
Abstract
Background The murine model of cerebral aneurysm (CA) serves as a prevalent tool for investigating the molecular underpinnings of CA. However, the extent to which the CA murine model aligns with that of human remains elusive. Methods The present study employed a comprehensive integration and exploration of the single-cell RNA-seq (scRNA-seq) datasets, along with multiple trajectory and gene regulatory network analyses, to investigate the cellular and molecular discrepancies between human and murine model CAs. Results The uniform manifold approximation and projection (umap) embedding exhibits that the primary discrepancies between human and murine model CAs reside in the cells of modifiable phenotype, encompassing vascular smooth muscle cell (vSMC), monocyte/macrophage, and neutrophil. The vSMCs from human CA tissue exhibit a fibroblast-like phenotype in comparison to that of murine model. Distinct patterns of neutrophil recruitment are observed in human and murine models, with the former characterized by neutrophil-derived CXCL8 and the latter by monocyte/macrophage-derived CCLs. In addition, macrophages originated from human unruptured CA express higher levels of M2 gene markers. Moreover, the inflammatory status of the CA tissue differs between humans and mouse models, with the former exhibiting a more acute and intense inflammation. Conclusion These findings demonstrate subtle but important disparities between human and murine model CAs, and may shed light upon an optimization of murine CA model.
Collapse
Affiliation(s)
- Hang Ji
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
- Department of Neurosurgery, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Guicheng Kuang
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Hailan Yang
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Haitao Liu
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Yue Li
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Shaoshan Hu
- Department of Neurosurgery, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Anqi Xiao
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Chao You
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Haogeng Sun
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Chaofeng Fan
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Guozhang Sun
- Department of Neurosurgery, Hei Longjiang Provincial People’s Hospital, Harbin, Hei Longjiang, China
| |
Collapse
|
38
|
Kou M, Xu Z, Guo Y, Zhang X, Wu M, Chen P, Liu Y, Tang X, Tang Y, Liu W. Development of a Mitochondria-Targeted Ruthenium(II)-Based Phosphorescent Probe for Hypochlorite Detection in Acute Inflammatory Model. Anal Chem 2025; 97:4987-4997. [PMID: 39993269 DOI: 10.1021/acs.analchem.4c05524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
The uncontrolled acute inflammatory response triggers dysregulation of the immunoinflammatory system, contributing to the development and progression of various acute inflammatory diseases (AIDs). Hypochlorite (ClO-), as a crucial oxidative mediator in AIDs, accumulates in the inflammatory environment, leading to direct cytotoxicity, secondary injury, and tissue dysfunction. However, achieving rapid detection, accurate tracking, in situ monitoring, and real-time imaging of ClO- in vivo remains a significant challenge. To address these issues, we developed a mitochondria-targeted phosphorescent probe (RuDM), which introduces a ligand containing a C═N bond as a ClO- recognition site to precisely identify ClO- in AIDs. It responds rapidly (6 s) and exhibits long-lived luminescence (471 ns), with a 190-fold luminescence enhancement in monitoring ClO-. Meanwhile, density functional theory (DFT) indicates that the luminescence enhancement of RuCOOH is attributed to the removal of an electron-withdrawing group (diaminomaleonitrile) from RuDM, which leads to an increase in the intersystem crossing rate and a greater probability of radiative transition from the T1 state. Finally, RuDM is used to monitor the levels of exogenous and endogenous ClO- in cells using confocal microscopy imaging and to evaluate its capability for ClO- detection over time in an acute inflammatory model. The above results suggest that RuDM, as a novel molecular platform to detect ClO-, has potential as a practical tool for research on the pathogenesis of acute inflammatory diseases.
Collapse
Affiliation(s)
- Manchang Kou
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Zhongsheng Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yanan Guo
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xinfeng Zhang
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Meilin Wu
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Peng Chen
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yun Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiaoliang Tang
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- Academy of Plateau Science and Sustainability, People's Government of Qinghai Province & Beijing Normal University, College of Chemistry and Chemical Engineering, Qinghai Normal University, Xining 810016, China
| | - Yu Tang
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Weisheng Liu
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- Academy of Plateau Science and Sustainability, People's Government of Qinghai Province & Beijing Normal University, College of Chemistry and Chemical Engineering, Qinghai Normal University, Xining 810016, China
| |
Collapse
|
39
|
Karjalainen A, Witalisz-Siepracka A, Prchal-Murphy M, Martin D, Sternberg F, Krunic M, Dolezal M, Fortelny N, Farlik M, Macho-Maschler S, Lassnig C, Meissl K, Amenitsch L, Lederer T, Pohl E, Gotthardt D, Bock C, Decker T, Strobl B, Müller M. Cell-type-specific requirement for TYK2 in murine immune cells under steady state and challenged conditions. Cell Mol Life Sci 2025; 82:98. [PMID: 40025196 PMCID: PMC11872851 DOI: 10.1007/s00018-025-05625-9] [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: 11/12/2024] [Revised: 01/31/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Tyrosine kinase 2 (TYK2) deficiency and loss or inhibition of kinase activity in men and mice leads to similar immune compromised phenotypes, predominantly through impairment of interferon (IFN) and interleukin 12 family responses. Here we relate the transcriptome changes to phenotypical changes observed in TYK2-deficient (Tyk2-/-) and TYK2 kinase-inactive (Tyk2K923E) mice in naïve splenic immune cells and upon ex vivo IFN treatment or in vivo tumor transplant infiltration. The TYK2 activities under homeostatic and both challenged conditions are highly cell-type-specific with respect to quantity and quality of transcriptionally dependent genes. The major impact of loss of TYK2 protein or kinase activity in splenic homeostatic macrophages, NK and CD8+ T cells and tumor-derived cytolytic cells is on IFN responses. While reportedly TYK2 deficiency leads to partial impairment of IFN-I responses, we identified cell-type-specific IFN-I-repressed gene sets completely dependent on TYK2 kinase activity. Reported kinase-inactive functions of TYK2 relate to signaling crosstalk, metabolic functions and cell differentiation or maturation. None of these phenotypes relates to respective enriched gene sets in the TYK2 kinase-inactive cell types. Nonetheless, the scaffolding functions of TYK2 are capable to change transcriptional activities at single gene levels and chromatin accessibility at promoter-distal regions upon cytokine treatment most prominently in CD8+ T cells. The cell-type-specific transcriptomic and epigenetic effects of TYK2 shed new light on the biology of this JAK family member and are relevant for current and future treatment of autoimmune and inflammatory diseases with TYK2 inhibitors.
Collapse
Affiliation(s)
- Anzhelika Karjalainen
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Agnieszka Witalisz-Siepracka
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems an Der Donau, Austria
| | - Michaela Prchal-Murphy
- Pharmacology and Toxicology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - David Martin
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Felix Sternberg
- Physiology and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Milica Krunic
- Campus Tulln, University of Applied Sciences Wiener Neustadt, Wiener Neustadt, Austria
| | - Marlies Dolezal
- Platform Biostatistics and Bioinformatics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Nikolaus Fortelny
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sabine Macho-Maschler
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Caroline Lassnig
- Core Facility VetBiomodels, University of Veterinary Medicine, Vienna, Austria
| | - Katrin Meissl
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Lena Amenitsch
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Therese Lederer
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Elena Pohl
- Physiology and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dagmar Gotthardt
- Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems an Der Donau, Austria
| | - Christoph Bock
- Cemm Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Thomas Decker
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna, Austria
| | - Birgit Strobl
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
40
|
Zhang K, Wang T, Huang X, Wu P, Shen L, Yang Y, Wan W, Sun S, Zhang Z. Ultrasound-mediated nanomaterials for the treatment of inflammatory diseases. ULTRASONICS SONOCHEMISTRY 2025; 114:107270. [PMID: 39961217 PMCID: PMC11875835 DOI: 10.1016/j.ultsonch.2025.107270] [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: 12/12/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
Sterile and infection-associated inflammatory diseases are becoming increasingly prevalent worldwide. Conventional drug therapies often entail significant drawbacks, such as the risk of drug overdose, the development of drug resistance in pathogens, and systemic adverse reactions, all of which can undermine the effectiveness of treatments for these conditions. Nanomaterials (NMs) have emerged as a promising tool in the treatment of inflammatory diseases due to their precise targeting capabilities, tunable characteristics, and responsiveness to external stimuli. Ultrasound (US), a non-invasive and effective treatment method, has been explored in combination with NMs to achieve enhanced therapeutic outcomes. This review provides a comprehensive overview of the recent advances in the use of US-mediated NMs for treating inflammatory diseases. A comprehensive introduction to the application and classification of US was first presented, emphasizing the advantages of US-mediated NMs and the mechanisms through which US and NMs interact to enhance anti-inflammatory therapy. Subsequently, specific applications of US-mediated NMs in sterile and infection-associated inflammation were summarized. Finally, the challenges and prospects of US-mediated NMs in clinical translation were discussed, along with an outline of future research directions. This review aims to provide insights to guide the development and improvement of US-mediated NMs for more effective therapeutic interventions in inflammatory diseases.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Xingyong Huang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Wenyu Wan
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, PR China; Key Laboratory of Immunodermatology, National Health Commission of the People's Republic of China, The First Hospital of China Medical University, PR China; National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, PR China.
| | - Siyu Sun
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China.
| | - Zhan Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, PR China; Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
41
|
Gorse L, Plessis L, Wearne S, Paradis M, Pinilla M, Chua R, Lim SS, Pelluz E, Toh GA, Mazars R, Bomfim C, Hervé F, Lhaute K, Réveillon D, Suire B, Ravon-Katossky L, Benoist T, Fromont L, Péricat D, Neil Mertens K, Derrien A, Terre-Terrillon A, Chomérat N, Bilien G, Séchet V, Carpentier L, Fall M, Sonko A, Hakim H, Sadio N, Bourdeaux J, Cougoule C, Henras AK, Perez-Oliva AB, Brehmer P, Roca FJ, Zhong FL, Common J, Meunier E, Hess P. Portimine A toxin causes skin inflammation through ZAKα-dependent NLRP1 inflammasome activation. EMBO Mol Med 2025; 17:535-562. [PMID: 39948420 PMCID: PMC11903881 DOI: 10.1038/s44321-025-00197-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 03/14/2025] Open
Abstract
In 2020-2021, a "mysterious illness" struck Senegalese fishermen, causing severe acute dermatitis in over one thousand individuals following exposure through drift-net fishing activity. Here, by performing deep analysis of the environmental samples we reveal the presence of the marine dinoflagellate Vulcanodinium rugosum and its associated cyclic imine toxins. Specifically, we show that the toxin PortimineA, strongly enriched in environmental samples, impedes ribosome function in human keratinocytes, which subsequently activates the stress kinases ZAKα and P38 and promotes the nucleation of the human NLRP1 inflammasome, leading to the release of IL-1β/IL-18 pro-inflammatory cytokines and cell death. Furthermore, cell-based models highlight that naturally occurring mutations in the P38-targeted sites of human NLRP1 are unable to respond to PortimineA exposure. Finally, the development and use of human organotypic skins and zebrafish models of PortimineA exposure demonstrate that the ZAKα-NLRP1 axis drives skin necrosis and inflammation. Our results exemplify the threats to human health caused by emerging environmental toxins and identify ZAKα and NRLP1 as important pharmacological targets to mitigate PortimineA toxicity.
Collapse
Affiliation(s)
- Léana Gorse
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Loïc Plessis
- Ifremer, PHYTOX Physiologie et Toxines des Microalgues Toxiques et Nuisibles, F-44000, Nantes, France
- Groupe Rocher, Research-Innovation & Development Department, Issy-les-Moulineaux, France
| | - Stephen Wearne
- A*STAR Skin Research, Institute of Singapore, Agency for Science, Technology and Research (A*STAR) Skin Research Labs, 138648, Singapore, Singapore
| | - Margaux Paradis
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Miriam Pinilla
- Department of Biochemistry and Molecular Biology-B and Immunology, Infectious Disease Pathology, Clinical Microbiology and Tropical Medicine, University of Murcia, Murcia, Spain
- Biomedical Research, Institute of Murcia (IMIB)-Pascual Parrilla, Murcia, Spain
| | - Rae Chua
- LKC School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Seong Soo Lim
- A*STAR Skin Research, Institute of Singapore, Agency for Science, Technology and Research (A*STAR) Skin Research Labs, 138648, Singapore, Singapore
| | - Elena Pelluz
- Department of Biochemistry and Molecular Biology-B and Immunology, Infectious Disease Pathology, Clinical Microbiology and Tropical Medicine, University of Murcia, Murcia, Spain
- Biomedical Research, Institute of Murcia (IMIB)-Pascual Parrilla, Murcia, Spain
| | - Gee-Ann Toh
- LKC School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Raoul Mazars
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Caio Bomfim
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Fabienne Hervé
- Ifremer, PHYTOX Physiologie et Toxines des Microalgues Toxiques et Nuisibles, F-44000, Nantes, France
| | - Korian Lhaute
- Ifremer, PHYTOX Physiologie et Toxines des Microalgues Toxiques et Nuisibles, F-44000, Nantes, France
| | - Damien Réveillon
- Ifremer, PHYTOX Physiologie et Toxines des Microalgues Toxiques et Nuisibles, F-44000, Nantes, France
| | - Bastien Suire
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Léa Ravon-Katossky
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Thomas Benoist
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Léa Fromont
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - David Péricat
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | | | | | | | | | | | - Véronique Séchet
- Ifremer, PHYTOX Physiologie et Toxines des Microalgues Toxiques et Nuisibles, F-44000, Nantes, France
| | - Liliane Carpentier
- Ifremer, PHYTOX Physiologie et Toxines des Microalgues Toxiques et Nuisibles, F-44000, Nantes, France
| | - Mamadou Fall
- Université Cheikh Anta Diop de Dakar, Laboratoire de Toxicologie et d'hydrologie, Dakar-Fann, Senegal
- Anti-Poison Centre, Fann University Hospital, Dakar, Senegal
| | - Amidou Sonko
- Anti-Poison Centre, Fann University Hospital, Dakar, Senegal
- Institut de Recherche pour le Développement, IRD, Univ Brest, CNRS, Ifremer, Dakar, Senegal
| | | | - Nfally Sadio
- Institut Sénégalais de Recherche Agricole, Centre de Recherche Océanographique de Dakar Thiaroye, Dakar, Senegal
| | - Jessie Bourdeaux
- Molecular, Cellular and Developmental (MCD) Unit, Centre for Integrative Biology (CBI), CNRS, University of Toulouse, UPS, Toulouse, France
| | - Céline Cougoule
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Anthony K Henras
- Molecular, Cellular and Developmental (MCD) Unit, Centre for Integrative Biology (CBI), CNRS, University of Toulouse, UPS, Toulouse, France
| | | | - Patrice Brehmer
- Institut de Recherche pour le Développement, IRD, Univ Brest, CNRS, Ifremer, Dakar, Senegal.
- SRFC, Sub regional Fisheries Commission, Liberté 5, Dakar, Senegal.
| | - Francisco J Roca
- Department of Biochemistry and Molecular Biology-B and Immunology, Infectious Disease Pathology, Clinical Microbiology and Tropical Medicine, University of Murcia, Murcia, Spain.
- Biomedical Research, Institute of Murcia (IMIB)-Pascual Parrilla, Murcia, Spain.
- Department of Biochemistry and Molecular Biology-B and Immunology, Infectious Disease Pathology, Clinical Microbiology and Tropical Medicine, University of Murcia, Murcia, Spain.
| | - Franklin L Zhong
- LKC School of Medicine, Nanyang Technological University, Singapore, Singapore.
- A*STAR Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR) Skin Research Labs, 138648, Singapore, Singapore.
| | - John Common
- A*STAR Skin Research, Institute of Singapore, Agency for Science, Technology and Research (A*STAR) Skin Research Labs, 138648, Singapore, Singapore.
- Translational and Clinical Research Institute and NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK.
- A*STAR Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR) Skin Research Labs, 138648, Singapore, Singapore.
| | - Etienne Meunier
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France.
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France.
| | - Philipp Hess
- Ifremer, PHYTOX Physiologie et Toxines des Microalgues Toxiques et Nuisibles, F-44000, Nantes, France.
- Ifremer, PHYTOX Physiologie et Toxines des Microalgues Toxiques et Nuisibles, F-44000, Nantes, France.
| |
Collapse
|
42
|
Fernandes DDO, Machado JR, Beltrami VA, Santos ACPMD, Queiroz-Junior CM, Vago JP, Soriani FM, Amaral FA, Teixeira MM, Felix FB, Pinho V. Disruption of survivin protein expression by treatment with YM155 accelerates the resolution of neutrophilic inflammation. Br J Pharmacol 2025; 182:1206-1222. [PMID: 39568085 DOI: 10.1111/bph.17375] [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: 01/20/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Prolonged survival of neutrophils is essential for determining the progression and severity of inflammatory and immune-mediated disorders, including gouty arthritis. Survivin, an anti-apoptotic molecule, has been described as a regulator of cell survival. This study aims to examine the effects of YM155 treatment, a survivin selective suppressant, in maintaining neutrophil survival in vitro and in vivo experimental settings of neutrophilic inflammation. EXPERIMENTAL APPROACH BALB/c mice were injected with monosodium urate (MSU) crystals and treated with YM155 (intra-articularly) at the peak of inflammatory response. Leukocyte recruitment, apoptosis neutrophil and efferocytosis were determined by knee joint wash cell morphology counting and flow cytometry. Resolution interval (Ri) was quantified by neutrophil infiltration, monitoring the amplitude and duration of the inflammation. Cytokine production was measured by ELISA. Mechanical hypernociception was assessed using an electronic von Frey aesthesiometer. Efferocytosis was evaluated in zymosan-induced neutrophilic peritonitis. Survivin and cleaved caspase-3 expression was determined in human neutrophils by flow cytometry. KEY RESULTS Survivin was expressed in neutrophils during MSU-induced gout, and the treatment with YM155 reduced survivin expression and shortened Ri from ∼8 h observed in vehicle-treated mice to ∼5.5 h, effect accompanied by increased neutrophil apoptosis and efferocytosis, both crucial for the inflammation resolution. Reduced IL-1β and CXCL1 levels were also observed in periarticular tissue. YM155 reduced histopathological score and hypernociceptive response. In human neutrophils, lipopolysaccharide (LPS) increased survivin expression, whereas survivin inhibition with YM155 induced neutrophil apoptosis, with activation of caspase-3. CONCLUSIONS AND IMPLICATIONS Survivin may be a promising therapeutic target to control neutrophilic inflammation.
Collapse
Affiliation(s)
- Débora de Oliveira Fernandes
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jessica Rayssa Machado
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vinicius Amorim Beltrami
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Priscila Vago
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frederico Marianetti Soriani
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávio Almeida Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Franciel Batista Felix
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
43
|
Lv R, Liu B, Jiang Z, Zhou R, Liu X, Lu T, Bao Y, Huang C, Zou G, Zhang Z, Lu L, Yin Q. Intermittent fasting and neurodegenerative diseases: Molecular mechanisms and therapeutic potential. Metabolism 2025; 164:156104. [PMID: 39674569 DOI: 10.1016/j.metabol.2024.156104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Neurodegenerative disorders are straining public health worldwide. During neurodegenerative disease progression, aberrant neuronal network activity, bioenergetic impairment, adaptive neural plasticity impairment, dysregulation of neuronal Ca2+ homeostasis, oxidative stress, and immune inflammation manifest as characteristic pathological changes in the cellular milieu of the brain. There is no drug for the treatment of neurodegenerative disorders, and therefore, strategies/treatments for the prevention or treatment of neurodegenerative disorders are urgently needed. Intermittent fasting (IF) is characterized as an eating pattern that alternates between periods of fasting and eating, requiring fasting durations that vary depending on the specific protocol implemented. During IF, depletion of liver glycogen stores leads to the production of ketone bodies from fatty acids derived from adipocytes, thereby inducing an altered metabolic state accompanied by cellular and molecular adaptive responses within neural networks in the brain. At the cellular level, adaptive responses can promote the generation of synapses and neurons. At the molecular level, IF triggers the activation of associated transcription factors, thereby eliciting the expression of protective proteins. Consequently, this regulatory process governs central and peripheral metabolism, oxidative stress, inflammation, mitochondrial function, autophagy, and the gut microbiota, all of which contribute to the amelioration of neurodegenerative disorders. Emerging evidence suggests that weight regulation significantly contributes to the neuroprotective effects of IF. By alleviating obesity-related factors such as blood-brain barrier dysfunction, neuroinflammation, and β-amyloid accumulation, IF enhances metabolic flexibility and insulin sensitivity, further supporting its potential in mitigating neurodegenerative disorders. The present review summarizes animal and human studies investigating the role and underlying mechanisms of IF in physiology and pathology, with an emphasis on its therapeutic potential. Furthermore, we provide an overview of the cellular and molecular mechanisms involved in regulating brain energy metabolism through IF, highlighting its potential applications in neurodegenerative disorders. Ultimately, our findings offer novel insights into the preventive and therapeutic applications of IF for neurodegenerative disorders.
Collapse
Affiliation(s)
- Renjun Lv
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Bin Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan 250014, China
| | - Ziying Jiang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Runfa Zhou
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehlstr. 13-17, Mannheim 68167, Germany
| | - Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Chunxia Huang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Guichang Zou
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Zongyong Zhang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China; National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871 Beijing, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
44
|
Li Y, Huang B, Yuan M, Zhang C, Zhang X, Hao J, Tao F, Geng F, Wang G, Su P. Associations between serum metal mixtures and systemic inflammation indices among Chinese early adolescents: A prospective cohort study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117952. [PMID: 40014987 DOI: 10.1016/j.ecoenv.2025.117952] [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: 12/17/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Research has demonstrated a link between metal exposure and inflammation. However, little is known about this relationship among adolescents, especially in prospective cohort studies. The aim of this study was to investigate the relationship between serum metal exposure and inflammatory status in Chinese early adolescents. METHODS In this study, 12 serum metals were detected at baseline in 1551 participants from the Chinese Early Adolescents Cohort. The participants' inflammatory status was assessed via three systemic inflammation indices (neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII)) at both baseline and follow-up. Generalized linear mixed models and restricted cubic splines regression were used to examine the linear and nonlinear relationships between single metal concentrations and systemic inflammation indices. Multiple mixture models were implemented to assess the relationships of mixed metals with systemic inflammation indices. Additionally, sex subgroup analyses were conducted to explore the sex-specific associations between serum metals and inflammatory status. RESULTS Single-exposure analysis revealed that exposure to multiple serum metals, such as chromium, cobalt, copper and lead, was positively associated with the NLR and SII, whereas iron was negatively correlated with the three systemic inflammation indices (PFDR<0.05). Additionally, inverted U-shaped associations were observed between vanadium, manganese and systemic inflammation indices. According to the mixture models, high levels of the serum metal mixture were positively correlated with the NLR and the SII. Cobalt had the highest positive weight in the mixed samples, whereas iron had the greatest negative weight in the serum-metal mixtures. Subgroup analyses revealed that serum exposure to the metal mixture had a more significant effect on systemic inflammation markers in females than in males. CONCLUSIONS This study reveals the impact of real-world mixed metal exposure on adolescents' inflammatory levels, which is of primary significance for protecting the healthy development of early adolescents.
Collapse
Affiliation(s)
- Yonghan Li
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China
| | - Binbin Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui 230032, China
| | - Mengyuan Yuan
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China
| | - Chao Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China
| | - Xueying Zhang
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jiahu Hao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui 230032, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui 230032, China
| | - Feng Geng
- Department of Psychology and Sleep Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China
| | - Gengfu Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui 230032, China.
| | - Puyu Su
- Department of Psychology and Sleep Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230001, China; School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China.
| |
Collapse
|
45
|
Hui L, Chen X, Huang M, Jiang Y, Liu T. TANK-Binding Kinase 1 in the Pathogenesis and Treatment of Inflammation-Related Diseases. Int J Mol Sci 2025; 26:1941. [PMID: 40076567 PMCID: PMC11900955 DOI: 10.3390/ijms26051941] [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: 01/13/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
TANK-binding kinase 1 (TBK1) is a key signaling kinase involved in innate immune and inflammatory responses. TBK1 drives immune cells to participate in the inflammatory response by activating the NF-κB and interferon regulatory factor signaling pathways in immune cells, promoting the expression of pro-inflammatory genes, and regulating immune cell function. Thus, it plays a crucial role in initiating a signaling cascade that establishes an inflammatory environment. In inflammation-related diseases, TBK1 acts as a bridge linking inflammation to immunity, metabolism, or tumorigenesis, playing an important role in the pathogenesis of immune-mediated inflammatory diseases, metabolic, inflammatory syndromes, and inflammation-associated cancers by regulating the activation of inflammatory pathways and the production of inflammatory cytokines in cells. In this review, we focused on the mechanisms of TBK1 in immune cells and inflammatory-related diseases, providing new insights for further studies targeting TBK1 as a potential treatment for inflammation-related diseases. Thus, optimizing and investigating highly selective cell-specific TBK1 inhibitors is important for their application in these diseases.
Collapse
Affiliation(s)
- Lu Hui
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
| | - Xiaolin Chen
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
| | - Mengke Huang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
| | - Yongmei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Ting Liu
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Biotherapy and Cancer Center/National Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
46
|
Jaschke NP, Wang A. Integrated control of leukocyte compartments as a feature of adaptive physiology. Immunity 2025; 58:279-294. [PMID: 39909034 DOI: 10.1016/j.immuni.2025.01.013] [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: 02/27/2024] [Revised: 12/10/2024] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
As a highly diverse and mobile organ, the immune system is uniquely equipped to participate in tissue responses in a tunable manner, depending on the number, type, and nature of cells deployed to the respective organ. Most acute organismal stressors that threaten survival-predation, infection, poisoning, and others-induce pronounced redistribution of immune cells across tissue compartments. Here, we review the current understanding of leukocyte compartmentalization under homeostatic and noxious conditions. We argue that leukocyte shuttling between compartments is a function of local tissue demands, which are linked to the organ's contribution to adaptive physiology at steady state and upon challenge. We highlight the neuroendocrine signals that relay and organize this trafficking behavior and outline mechanisms underlying the functional diversification of leukocyte responses. In this context, we discuss important areas of future inquiry and the implications of this scientific space for clinical medicine in the era of targeted immunomodulation.
Collapse
Affiliation(s)
- Nikolai P Jaschke
- Department of Internal Medicine (Rheumatology, Allergy & Immunology) and Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
| | - Andrew Wang
- Department of Internal Medicine (Rheumatology, Allergy & Immunology) and Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
47
|
Huang X, Kuang M, Qiu J, Wang C, Sheng G, Zou Y, Xie G. Assessment of platelet-to-white blood cell ratio on short-term mortality events in patients hospitalized with acute decompensated heart failure: evidence from a cohort study from Jiangxi, China. Front Cardiovasc Med 2025; 12:1454933. [PMID: 39991636 PMCID: PMC11842369 DOI: 10.3389/fcvm.2025.1454933] [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: 06/27/2024] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
Objective Platelet-to-white blood cell ratio (PWR) as a comprehensive indicator of inflammatory response has been widely used to assess the prognosis of various diseases. However, the relationship between PWR and adverse outcomes in patients with acute decompensated heart failure (ADHF) remains unclear. This study aimed to evaluate the association between PWR and all-cause mortality within 30 days of hospitalization in ADHF patients from Jiangxi, China. Methods A total of 1,453 ADHF patients from the Jiangxi-ADHF study1 cohort were included. The primary outcome measure was all-cause mortality within 30 days of hospitalization. Multivariable Cox proportional hazards regression, restricted cubic spline regression, and receiver operating characteristic curve analysis were employed to explore the association between the inflammatory marker PWR and all-cause mortality in ADHF patients within 30 days of hospitalization. Results During the 30-day observation period, a total of 53 subjects experienced mortality events. Multivariable Cox regression showed a negative correlation between PWR and all-cause mortality within 30 days of hospitalization in ADHF patients. Restricted cubic spline regression demonstrated an L-shaped association between PWR and 30-day mortality risk (p for nonlinear = 0.038). Further threshold analysis revealed a threshold point for PWR at 15.88, where a decrease in PWR below this threshold was significantly associated with increased risk of all-cause mortality (p for log-likelihood ratio test = 0.046). Additionally, the results of receiver operating characteristic curve analysis indicated that PWR had high predictive accuracy for mortality events within 30 days of hospitalization in ADHF patients and is significantly better than the traditional HF marker N-Terminal Pro-Brain Natriuretic Peptide (AUC: NT-proBNP 0.69, PWR 0.76; Delong test P < 0.05). Subgroup analysis showed that compared to subjects with reduced or moderately reduced ejection fraction, ADHF patients with preserved ejection fraction had a lower risk of short-term mortality associated with PWR (HR:0.99 vs. 0.98 vs. 0.87, P for interaction = 0.0067). Conclusion This study reveals, for the first time, a negative correlation between the inflammatory marker PWR and all-cause mortality within 30 days of hospitalization in ADHF patients. Based on the threshold analysis findings, patients with ADHF and a PWR below 15.88 had a significantly higher risk of death within 30 days.
Collapse
Affiliation(s)
- Xin Huang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Maobin Kuang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Jiajun Qiu
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Chao Wang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Guotai Sheng
- Jiangxi Provincial Geriatric Hospital, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Yang Zou
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Guobo Xie
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
48
|
Rai P, Fessler MB. Mechanisms and effects of activation of innate immunity by mitochondrial nucleic acids. Int Immunol 2025; 37:133-142. [PMID: 39213393 DOI: 10.1093/intimm/dxae052] [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: 07/10/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
In recent years, a growing number of roles have been identified for mitochondria in innate immunity. One principal mechanism is that the translocation of mitochondrial nucleic acid species from the mitochondrial matrix to the cytosol and endolysosomal lumen in response to an array of microbial and non-microbial environmental stressors has been found to serve as a second messenger event in the cell signaling of the innate immune response. Thus, mitochondrial DNA and RNA have been shown to access the cytosol through several regulated mechanisms involving remodeling of the mitochondrial inner and outer membranes and to access lysosomes via vesicular transport, thereby activating cytosolic [e.g. cyclic GMP-AMP synthase (cGAS), retinoic acid-inducible gene I (RIG-I)-like receptors], and endolysosomal (Toll-like receptor 7, 9) nucleic acid receptors that induce type I interferons and pro-inflammatory cytokines. In this mini-review, we discuss these molecular mechanisms of mitochondrial nucleic acid mislocalization and their roles in host defense, autoimmunity, and auto-inflammatory disorders. The emergent paradigm is one in which host-derived DNA interestingly serves as a signal amplifier in the innate immune response and also as an alarm signal for disturbances in organellar homeostasis. The apparent vast excess of mitochondria and mitochondrial DNA nucleoids per cell may thus serve to sensitize the cell response to stressors while ensuring an underlying reserve of intact mitochondria to sustain cellular metabolism. An improved understanding of these molecular mechanisms will hopefully afford future opportunities for therapeutic intervention in human disease.
Collapse
Affiliation(s)
- Prashant Rai
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
49
|
Llorca T, Ruiz-Magaña MJ, Abadía AC, Ruiz-Ruiz C, Olivares EG. Decidual stromal cells: fibroblasts specialized in immunoregulation during pregnancy. Trends Immunol 2025; 46:138-152. [PMID: 39947975 DOI: 10.1016/j.it.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025]
Abstract
Decidual stromal cells (DSCs) are involved in immunoregulatory mechanisms that prevent fetal rejection by the mammalian maternal immune system. Recent studies using single-cell RNA sequencing demonstrated the existence of different types of human and mouse DSCs, highlighting corresponding differentiation (decidualization) pathways, and suggesting their involvement in the immune response during normal and pathological pregnancy. DSCs may be considered tissue-specialized fibroblasts because both DSCs and fibroblasts share phenotypic and functional similarities in immunologically challenged tissues, especially in terms of their immune functions. Indeed, fibroblasts can setup, support, and suppress immune responses and these functions are also performed by DSCs. Moreover, fibroblasts and DSCs can induce ectopic foci as tertiary lymphoid structures (TLSs), and endometriosis, respectively. Thus, understanding DSC immunoregulatory functions is of timely relevance.
Collapse
Affiliation(s)
- Tatiana Llorca
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain
| | - María José Ruiz-Magaña
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Biología Celular, Universidad de Granada, Granada, Spain.
| | - Ana C Abadía
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Carmen Ruiz-Ruiz
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Enrique G Olivares
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain.
| |
Collapse
|
50
|
Qi Y, Li C, Gao X, Zhang F. Causal relationship between circulating inflammatory cytokines and the risk of hernia: a bidirectional Mendelian randomization study. J Int Med Res 2025; 53:3000605251315923. [PMID: 39956620 PMCID: PMC11831628 DOI: 10.1177/03000605251315923] [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: 09/22/2024] [Accepted: 01/09/2025] [Indexed: 02/18/2025] Open
Abstract
OBJECTIVE Observational studies suggest a link between hernia and inflammatory cytokines, but randomized trials are limited by ethical and cost constraints. In this study, we used bidirectional Mendelian randomization (MR) to investigate the causal relationship between inflammatory cytokines and five types of hernia, aiming to inform preventive and therapeutic strategies. METHODS We selected 41 inflammatory factors and five types of hernia as instrumental variables, using data from the IEU Open GWAS database including individuals of European descent. The primary analysis used the inverse variance weighted method with false discovery rate (FDR) adjustment. Additional MR methods and sensitivity analyses ensured robustness. Reverse MR was used to assess potential reverse causality. RESULTS After FDR adjustment, stem cell growth factor beta (SCGFb) was causally associated with diaphragmatic hernia (odds ratio = 0.884, 95% confidence interval: 0.819-0.955). Reverse MR indicated that diaphragmatic hernia may increase interferon gamma-induced protein 10 (IP10) and monokine induced by interferon-gamma (MIG), and ventral hernia may elevate macrophage inflammatory protein-1b (MIP1b). Sensitivity analyses confirmed robustness. CONCLUSION SCGFb may protect against diaphragmatic hernia, and IP10, MIG, and MIP1b are involved in hernia development, suggesting the therapeutic potential of targeting these cytokines. Further studies are needed.
Collapse
Affiliation(s)
- Yaqin Qi
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People’s Hospital, Hangzhou, China
| | - Changjiu Li
- Department of Urology, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Xingyue Gao
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou Third People’s Hospital, Hangzhou, China
| | - Fangjie Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People’s Hospital, Hangzhou, China
| |
Collapse
|