1
|
Xia Q, Huang X, Li A, Zhuang Z, Zhou X, Yang Y, Zuo X, Liu Y, Sheng Y, Xu J, Cui Y. OASL activates MAPK to drive psoriatic pathogenesis: Astilbin targeting this axis improves metabolic-inflammation crosstalk. Life Sci 2025; 375:123698. [PMID: 40360089 DOI: 10.1016/j.lfs.2025.123698] [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: 02/19/2025] [Revised: 04/23/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
AIMS This study aimed to elucidate the role of oligoadenylate synthase-like protein (OASL) as a pivotal regulator integrating keratinocyte hyperproliferation, inflammation, and lipid metabolic dysregulation in psoriasis pathogenesis. MATERIALS AND METHODS Clinical analyses compared epidermal OASL expression levels between psoriasis patients and healthy individuals. Functional studies in HaCaT cells employed OASL knockdown and overexpression to assess effects on proliferation, inflammatory responses, and lipid metabolism. The JAK1-STAT1-OASL regulatory axis was investigated using the JAK1 inhibitor Upadacitinib. The natural flavonoid Astilbin was screened as an OASL inhibitor, and its therapeutic efficacy was evaluated in imiquimod-induced psoriatic mice. KEY FINDINGS OASL was significantly upregulated in psoriatic epidermis. Knockdown of OASL suppressed keratinocyte proliferation and inflammation, while its overexpression promoted hyperproliferation, inflammation, and lipid metabolic dysregulation via p38 MAPK pathway activation. Mechanistically, Upadacitinib reduced OASL expression by inhibiting STAT1, confirming the JAK1-STAT1-OASL axis. Astilbin markedly alleviated imiquimod-induced psoriasiform dermatitis in mice. SIGNIFICANCE This study reveals OASL's pivotal regulatory role in psoriasis and its associated pathways, providing novel therapeutic targets and a potential natural drug candidate. These findings establish a theoretical foundation for developing multi-targeted strategies against psoriasis.
Collapse
Affiliation(s)
- Qingyue Xia
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China.
| | - Xiaoyi Huang
- Institute of Dermatology and Department of Dermatology of the First Affiliated Hospital, Anhui Medical University, Hefei 230031, China
| | - Ang Li
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
| | - Zhou Zhuang
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xinzhu Zhou
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Yue Yang
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Institute of Clinical Medical Sciences, Beijing 100029, China
| | - Xianbo Zuo
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yi Liu
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; Capital Medical University, Beijing 100069, China
| | - Yujun Sheng
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; Institute of Dermatology and Department of Dermatology of the First Affiliated Hospital, Anhui Medical University, Hefei 230031, China.
| | - Jingkai Xu
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Yong Cui
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
2
|
Qin L, Ji X, Zhu S, Zhu Z, Yang S, Li H, Bai L, Yu Z, Li X. Astilbin ameliorates intracerebral hemorrhage-induced secondary brain injury by upregulating Treg cells in mice. Biochem Biophys Res Commun 2025; 763:151805. [PMID: 40233435 DOI: 10.1016/j.bbrc.2025.151805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/31/2025] [Accepted: 04/10/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Intracerebral Hemorrhage (ICH) is a common acute fatal cerebrovascular disease in surgery. Astilbin, a flavonoid extracted from various plants, has been studied for its excellent anti-inflammatory action. The present study aims to investigate the efficacy and mechanism of astilbin against ICH in mice. METHODS The ICH in C57BL/6 mice was induced with classical autologous blood injection. We conducted behavioral tests, Perls' staining, Nissl staining, TUNEL staining, Evans blue dye extravasation, water content, Enzyme-linked immunosorbent assay (ELISA), immunofluorescence staining to access the anti-inflammatory function of astilbin. RESULTS Astilbin demonstrates a capacity to ameliorate neurofunctional impairments induced by ICH in mice. It effectively reduces iron deposition and neuronal death in the peri-hematoma tissue. Astilbin alleviates cerebral edema and mitigates blood-brain barrier (BBB) damage caused by ICH. Furthermore, astilbin promotes the secretion of chemotactic and anti-inflammatory factors post-ICH, including CCL1, CCL20, IL-10, IL-35, and TGFβ. Notably, astilbin facilitates the accumulation of Treg cells in the brain tissue surrounding the hematoma after ICH. CONCLUSION Astilbin facilitates the enrichment of Treg cells in the brain tissue around the hematoma, thereby alleviating secondary brain injury (SBI) following ICH and improving the overall prognosis of ICH. These findings suggest its potential candidacy as a therapeutic intervention for mitigating the consequences of intracerebral hemorrhage.
Collapse
Affiliation(s)
- Lei Qin
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Xiaoyu Ji
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Shixin Zhu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Zhenghua Zhu
- Department of Clinical Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Siyuan Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Lei Bai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China.
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
3
|
Kim JW, Tung HC, Yang B, Pant R, Guan X, Feng Y, Xie W. Heme-thiolate monooxygenase cytochrome P450 1B1, an old dog with many new tricks. Pharmacol Rev 2025; 77:100045. [PMID: 40054133 DOI: 10.1016/j.pharmr.2025.100045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 05/12/2025] Open
Abstract
Cytochrome P450 CYP1B1 is a heme-thiolate monooxygenase traditionally recognized for its xenobiotic functions and extrahepatic expressions. Recent studies have suggested that CYP1B1 is also expressed in hepatic stellate cells, immune cells, endothelial cells, and fibroblasts within the tumor microenvironment, as well as tumor cells themselves. CYP1B1 is responsible for the metabolism of a wide range of substrates, including xenobiotics such as drugs, environmental chemicals, and endobiotics such as steroids, retinol, and fatty acids. Consequently, CYP1B1 and its associated exogenous and endogenous metabolites have been critically implicated in the pathogenesis of many diseases. Understanding the mode of action of CYP1B1 in different pathophysiological conditions and developing pharmacological inhibitors that allow for systemic or cell type-specific modulation of CYP1B1 may pave the way for novel therapeutic opportunities. This review highlights the significant role of CYP1B1 in maintaining physiological homeostasis and provides a comprehensive discussion of recent advancements in our understanding of CYP1B1's involvement in the pathogenesis of diseases such as fibrosis, cancer, glaucoma, and metabolic disorders. Finally, the review emphasizes the therapeutic potential of targeting CYP1B1 for drug development, particularly in the treatment and prevention of cancers and liver fibrosis. SIGNIFICANCE STATEMENT: CYP1B1 plays a critical role in various physiological processes. Dysregulation or genetic mutations of the gene encoding this enzyme can lead to health complications and may increase the risk of diseases such as cancer and liver fibrosis. In this review, we summarize recent preclinical and clinical evidence that underscores the potential of CYP1B1 as a therapeutic target.
Collapse
Affiliation(s)
- Jong-Won Kim
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hung-Chun Tung
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bin Yang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rajat Pant
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiuchen Guan
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Ye Feng
- Department of Endocrinology and Metabolic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
4
|
Zhang BH, Chen H, Yang R, Jiang Z, Huang S, Chen Z, Liu C, Wang L, Liu XH. Pinocembrin alleviates renal ischemia-reperfusion injury/unilateral ureteral obstruction (UUO)-generated renal fibrosis by targeting the CYP1B1/ROS/MAPK axis. FEBS J 2025; 292:2119-2144. [PMID: 39876048 DOI: 10.1111/febs.17414] [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: 07/01/2024] [Revised: 10/23/2024] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
Abstract
In our research, we constructed models of renal ischemia-reperfusion (I/R)-exposed acute kidney injury (AKI) and unilateral ureteral obstruction (UUO)-stimulated renal fibrosis (RF) in C57BL/6 mice and HK-2 cells. We firstly authenticated that oral pinocembrin (PIN) administration obviously mitigated tissue damage and renal dysfunction induced by I/R injury, and PIN attenuated UUO-caused RF, as confirmed by the reduced expression of fibrotic markers as well as hematoxylin-eosin (H&E), Sirius red, immunohistochemistry, and Masson staining. Meanwhile, the beneficial role of PIN was again demonstrated in HK-2 cells with hypoxia-reoxygenation (H/R) or transforming growth factor beta-1 (TGF-β1) treatment. Importantly, the "ingredient-target-pathway-disease" network was established through bioinformatics analysis and molecular docking, which showed that PIN may target cytochrome P450 1B1 (CYP1B1) and modulate the mitogen-activated protein kinase (MAPK) pathway to exert its impact during injury. Furthermore, experiments confirmed that PIN usage remarkably constrained CYP1B1 expression, reactive oxygen species (ROS) production, MAPK-pathway-associated inflammation, or apoptosis during I/R injury or UUO exposure. PIN also ameliorated the elevated protein phosphorylation of MAPK pathway components [p38, extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase 1 (JNK ERK and JNK)], which validated the PIN-induced inhibition of the MAPK signaling pathway in renal I/R or UUO injury. Moreover, the AAV9 (adeno-associated virus 9)-packed CYP1B1 or pcDNA-CYP1B1 overexpression plasmid was utilized to treat C57BL/6 mice or HK-2 cells to overexpress CYP1B1, respectively. Notably, CYP1B1 overexpression considerably abolished PIN's restriction impact on ROS generation and MAPK pathway activation. In conclusion, via bioinformatics analysis, molecular docking, animal model, and cellular experiments, we proved that PIN alleviates renal I/R injury/UUO-generated renal fibrosis through regulating the CYP1B1/ROS/MAPK axis.
Collapse
Affiliation(s)
- Bang-Hua Zhang
- Department of Urology, Renmin Hospital of Wuhan University, China
- Wuhan University Institute of Urological Disease, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, China
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hui Chen
- Department of Urology, Renmin Hospital of Wuhan University, China
- Wuhan University Institute of Urological Disease, China
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, China
- Wuhan University Institute of Urological Disease, China
| | - Zhengyu Jiang
- Department of Urology, Renmin Hospital of Wuhan University, China
- Wuhan University Institute of Urological Disease, China
| | - Shiyu Huang
- Department of Urology, Renmin Hospital of Wuhan University, China
- Wuhan University Institute of Urological Disease, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, China
- Wuhan University Institute of Urological Disease, China
| | - Cheng Liu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, China
- Wuhan University Institute of Urological Disease, China
| | - Xiu-Heng Liu
- Department of Urology, Renmin Hospital of Wuhan University, China
- Wuhan University Institute of Urological Disease, China
| |
Collapse
|
5
|
Shi Z, Liu J, Qin J, Liang X, Ou X, Zhang T, Yan X, Hu Q, Huang W, Hu K. Astilbin Alleviates Radiation-Induced Pulmonary Fibrosis via circPRKCE Targeting the TGF-β/Smad7 Pathway to Inhibit Epithelial-Mesenchymal Transition. Biomedicines 2025; 13:689. [PMID: 40149664 PMCID: PMC11939908 DOI: 10.3390/biomedicines13030689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
Purpose: This study aimed to clarify the protective effect of astilbin (AST) on radiation-induced pulmonary fibrosis (RIPF) and explore its underlying molecular mechanism, focusing on non-coding RNAs. Methods: Mouse lung epithelial cells (MLE-12 and TC-1) and C57BL/6J mice were used to establish in vitro radiation injury models and in vivo RIPF models, respectively. Cell viability, apoptosis, the epithelial-to-mesenchymal transition (EMT), and fibrosis-related markers were assessed using cell-counting kit-8 assays, Western blotting, immunohistochemistry, and histological staining. High-throughput sequencing identified differentially expressed circRNAs. The mechanistic studies included RNA-FISH, a dual-luciferase reporter assay, an RNA immunoprecipitation (RIP) assay, and loss-of-function experiments. Results: AST significantly alleviated radiation-induced apoptosis and EMT in vitro, as well as RIPF in vivo. AST treatment reduced collagen deposition, fibrosis-related protein expression, and EMT marker changes. High-throughput sequencing revealed that AST upregulated circPRKCE, a non-coding RNA that functions through a ceRNA mechanism by binding to miR-15b-5p, thereby promoting Smad7 expression and suppressing the TGF-β/Smad7 pathway. Knockdown of circPRKCE abolished AST's protective effects, confirming its pivotal role in mediating AST's anti-fibrotic activity. Conclusions: This study demonstrates that Astilbin alleviates radiation-induced pulmonary fibrosis via circPRKCE targeting the TGF-β/Smad7 pathway to inhibit EMT, suggesting AST as a potential therapeutic agent for managing this severe complication of radiotherapy.
Collapse
Affiliation(s)
- Zhiling Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Jing Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Jing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Xian Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Xue Ou
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China
| | - Tingting Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Xueting Yan
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Qianxin Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Weimei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Kai Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
6
|
Saikia L, Talukdar NC, Dutta PP. Exploring the Therapeutic Role of Flavonoids Through AMPK Activation in Metabolic Syndrome: A Narrative Review. Phytother Res 2025; 39:1403-1421. [PMID: 39789806 DOI: 10.1002/ptr.8428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 01/12/2025]
Abstract
Metabolic syndrome (MetS) is a cluster of interrelated metabolic abnormalities that significantly elevate the risk of cardiovascular disease, obesity, and diabetes. Flavonoids, a diverse class of bioactive polyphenolic compounds found in plant-derived foods and beverages, have garnered increasing attention as potential therapeutic agents for improving metabolic health. This review provides a comprehensive analysis of the therapeutic effects of flavonoids in the context of the MetS, with a particular focus on their modulation of the AMP-activated protein kinase (AMPK) pathway. AMPK serves as a central regulator of cellular energy balance, glucose metabolism, and lipid homeostasis, making it a critical target for metabolic intervention. Through a systematic review of the literature up to April 2024, preclinical studies across various flavonoid subclasses, including flavonols, and flavan-3-ols, were analysed to elucidate their mechanistic roles in metabolic regulation. Many studies suggests that flavonoids enhance glycolipid metabolism by facilitating glucose transporter 4 (GLUT4) translocation and activating the AMPK pathway, thereby improving glycemic control in diabetes models. In obesity-related studies, flavonoids demonstrated significant inhibitory effects on lipid synthesis, reduced adipogenesis, and attenuated proinflammatory cytokine secretion via AMPK activation. These findings show the broad therapeutic potential of flavonoids in addressing the MetS and its associated disorders. While these preclinical insights highlight flavonoids as promising natural agents for metabolic health improvement, it is important to note that their excessive concentrations may disrupt these pathways, potentially leading to metabolic imbalance and cytotoxicity. Further studies and clinical trials are essential to determine optimal dosing regimens, formulations, and the long-term safety and efficacy of flavonoids. This review highlights the importance of flavonoids for natural interventions targeting MetS and its comorbidities, offering a foundation for future translational research.
Collapse
Affiliation(s)
- Lunasmrita Saikia
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, India
| | | | - Partha Pratim Dutta
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, India
| |
Collapse
|
7
|
Lai L, Wu H, Peng L, Zhang Z, Wu X, Zheng S, Su Z, Chu H. GelMA@LNP/AST Promotes eNOS-Dependent Angiogenesis Through Autophagy Activation for the Treatment of Hind Limb Ischemia. Int J Nanomedicine 2025; 20:1821-1841. [PMID: 39958322 PMCID: PMC11829644 DOI: 10.2147/ijn.s499478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/28/2025] [Indexed: 02/18/2025] Open
Abstract
Purpose Limb ischemia is a refractory disease characterized by insufficient angiogenesis and tissue necrosis. Currently, the primary clinical treatment method is surgical intervention; however, the prognosis for patients with severe limb ischemia remains unsatisfactory. Although some studies have evaluated the effects of using bioactive factors to promote neovascularization and tissue repair, the clinical outcomes have not met expectations, possibly due to the difficulties in maintaining biological activity and avoiding potential side effects. Traditional Chinese medicine, specifically astilbin (AST), is a potential therapeutic agent in promoting tissue regeneration. However, there have been no reports on its efficacy in treating limb ischemia through promoting angiogenesis. Materials and Methods In this study, we prepared AST-loaded lignin nanoparticles (LNP/AST) with sustained-release functionality, which were mixed with GelMA hydrogel (GelMA@LNP/AST). The angiogenic effects were evaluated in a mouse model of hind limb ischemia. To further investigate the mechanism of angiogenesis, human endothelial cell line EA.hy926 was exposed to different concentrations of AST. The effects of AST on cell migration and angiogenesis were studied using wound healing assays and angiogenesis assays. The changes in angiogenesis markers, autophagy markers, and eNOS levels were detected using qPCR and Western blotting. 3-MA was used to assess the role of autophagy in the activation of eNOS mediated by AST and its subsequent angiogenic effects. Results GelMA@LNP/AST significantly promoted blood flow recovery in mice with hind limb ischemia. This effect was mainly attributed to the enhanced migration and angiogenic capabilities of endothelial cells mediated by AST. A potential underlying mechanism could be that the autophagy induced by AST increases eNOS activity. Conclusion GelMA@LNP/AST enables complete revascularization in female mice after hind limb ischemia, thereby achieving limb preservation and restoring motor function. Given the good therapeutic potential of the GelMA@LNP/AST in revascularization, it may become an effective strategy for successfully salvaging limbs in cases of limb ischemia.
Collapse
Affiliation(s)
- Lingzhi Lai
- Maoming People’s Hospital, Maoming, Guangdong, People’s Republic of China
| | - Hao Wu
- Maoming People’s Hospital, Maoming, Guangdong, People’s Republic of China
| | - Liang Peng
- The First People’s Hospital of Guiyang, Guiyang, Guizhou, People’s Republic of China
| | - Zhen Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Xinfan Wu
- Maoming People’s Hospital, Maoming, Guangdong, People’s Republic of China
| | - Shuo Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Zekang Su
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Hongxing Chu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
8
|
Huang R, Lu J, Yang X, Sheng G, Qin F, Yang X. Molecular mechanism of the effect of BixiezelanYin on knee osteoarthritis based on network pharmacology and molecular docking. Medicine (Baltimore) 2025; 104:e41459. [PMID: 39928767 PMCID: PMC11813014 DOI: 10.1097/md.0000000000041459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 02/12/2025] Open
Abstract
The BixizelanYin (BXZLY) is a traditional herbal formula for treating damp-heat type knee osteoarthritis (KOA). This study aimed to investigate the potential therapeutic effects of BXZLY on KOA by using network pharmacology and molecular docking. Active ingredients and their corresponding targets of BXZLY were screened through the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform. The known disease targets of KOA were retrieved from GeneCards and the Online Mendelian Inheritance in Man database. The effective targets of BXZLY for treating KOA were obtained with the Venny platform. A drug-component-target network diagram was constructed using Cytoscape 3.7.2 software, and a protein-protein interaction network was established via the STRING database. The protein-protein interaction network was visualized by Cytoscape 3.7.2 software. The biological functions of the targets wereanalyzed through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis with the DAVID database. The Gene Ontology enrichment and KEGG pathway visualization bubble charts were generated using the OmicShare online platform to explore the biological functions of BXZLY in treating KOA. The main signaling pathways of BXZLY in treating KOA were investigated by KEGG pathway enrichment analysis. Finally, AutoDockTools software was used for molecular docking validation of the key molecules of the drug and the key proteins of the disease. A total of 108 active compounds and 280 target proteins of BXZLY were identified. There were 2816 disease-related targets related to KOA, among which 99 were targeted by BXZLY for KOA treatment. Key compounds included β-sitosterol, stigmasterol, campesterol, quercetin, and dioscin. Core target proteins consisted of STAT3, ESR1, EGFR, JUN, and PTGS2. The pathways mainly involved the advanced glycation end products - receptor for advanced glycation end products, estrogen, HIF-1, and relaxin signal pathways. Molecular docking showed a strong binding affinity between active ingredients and core target proteins. Docking candidates were selected based on oral bioavailability ≥ 30%, drug-likeness ≥ 0.18, and degree values in Cytoscape. This strategy ensured a reliable selection of compounds with high therapeutic relevance. BXZLY provides a comprehensive therapeutic strategy for KOA treatment by regulating cell proliferation and alleviating inflammation through its multi-component, multi-target, and multi-pathway effects.
Collapse
Affiliation(s)
- Renkun Huang
- Guangxi University of Chinese Medicine, Nanning City, Guangxi, China
- Liuzhou Traditional Chinese Medicine Hospital, Liuzhou City, Guangxi, China
| | - Jiehua Lu
- Guangxi University of Chinese Medicine, Nanning City, Guangxi, China
- Ruikang Hospital, Affiliated to Guangxi University of Chinese Medicine, Nanning City, Guangxi, China
| | - Xueyi Yang
- Liuzhou Traditional Chinese Medicine Hospital, Liuzhou City, Guangxi, China
| | - Guanyun Sheng
- Liuzhou Traditional Chinese Medicine Hospital, Liuzhou City, Guangxi, China
| | - Fangyi Qin
- Guangxi University of Chinese Medicine, Nanning City, Guangxi, China
| | - Xiongwu Yang
- Liuzhou Traditional Chinese Medicine Hospital, Liuzhou City, Guangxi, China
| |
Collapse
|
9
|
Wang S, Xu K, Du W, Gao X, Ma P, Yang X, Chen M. Exposure to environmental doses of DEHP causes phenotypes of polycystic ovary syndrome. Toxicology 2024; 509:153952. [PMID: 39265699 DOI: 10.1016/j.tox.2024.153952] [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: 07/27/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Globally, approximately 6-20 % of women who are of reproductive age suffer from polycystic ovary syndrome (PCOS), with environmental factors believed to be significant contributors. Di-2-ethylhexyl phthalate (DEHP) is known to be an endocrine disruptor, and is also suspected of being associated with the occurrence of PCOS, but in vivo studies to verify this association are lacking. In this study, female SD rats were exposed to DEHP at levels of 0.1, 1.0, and 10 mg/kg/d, which are comparable to daily human exposure, to explore its potential role in the development of PCOS. The findings indicated that DEHP exposure reduced ovarian and uterine coefficients, decreased accumulation of primordial follicles, increased the prevalence of atretic and cystic follicles and fibrosis in ovarian tissues, altered serum hormone levels, elevated blood glucose levels and insulin resistance, disrupted the endocrine system and resulted in significant oxidative damage in the ovarian tissues. These results imply that DEHP exposure may cause lesions resembling PCOS to develop. By analyzing the differential expression of the proteome, and using GO and KEGG enrichment analyses, we found they were mainly enriched in the metabolic pathway and in the PPAR signaling pathway. We confirmed that activation of the PPARγ signaling pathway caused by DEHP exposure, is related to the emergence of PCOS-like lesions. This research provides direct in vivo experimental evidence for the association between DEHP exposure and PCOS.
Collapse
Affiliation(s)
- Shuxin Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei 430079, China
| | - Ke Xu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei 430079, China
| | - Wanting Du
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei 430079, China
| | - Xiao Gao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei 430079, China
| | - Ping Ma
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Xu Yang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei 430079, China; Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Mingqing Chen
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, Hubei 430079, China.
| |
Collapse
|
10
|
Dhiman S, Dhankhar S, Garg A, Rohilla M, Saini M, Singh TG, Chauhan S, Selim S, Al Jaouni SK, Yasmin S, Begum N, Alshahrani A, Ansari MY. Mechanistic insights and therapeutic potential of astilbin and apigenin in diabetic cardiomyopathy. Heliyon 2024; 10:e39996. [PMID: 39583813 PMCID: PMC11582444 DOI: 10.1016/j.heliyon.2024.e39996] [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: 09/27/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) represents a critical complication of Diabetes mellitus (DM), characterized by structural and functional changes in the myocardium independent of coronary artery disease or hypertension. Emerging evidence highlights the significant roles of phytochemicals, particularly astilbin and apigenin, in modulating key molecular pathways implicated in DCM. This review synthesizes current mechanistic insights and therapeutic potential of these compounds, focusing on their interactions with AMP-activated protein kinase (AMPK), peroxisome proliferator-activated receptors (PPARs), O-linked N-acetylglucosamine (O-GlcNAc), sodium-glucose co-transporter 2 (SGLT2), protein kinase C (PKC), nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (MAPK), and c-Jun N-terminal kinase (JNK) pathways. Astilbin and apigenin have demonstrated the ability to improve cardiac function, mitigate oxidative stress, and reduce inflammatory responses in diabetic conditions. By activating AMPK and PPARs, these flavonoids enhance glucose uptake and fatty acid oxidation, contributing to improved metabolic homeostasis. Their inhibition of O-GlcNAcylation, SGLT2 activity, and PKC signaling further attenuates hyperglycemia-induced cellular damage. Additionally, suppression of NF-κB, MAPK, and JNK pathways by astilbin and apigenin results in reduced pro-inflammatory cytokine production and apoptotic cell death. Collectively, these interactions position astilbin and apigenin as promising therapeutic agents for ameliorating DCM, offering novel avenues for treatment strategies aimed at modulating multiple pathogenic pathways.
Collapse
Affiliation(s)
- Sachin Dhiman
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Sanchit Dhankhar
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Anjali Garg
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Swami Devi Dyal College of Pharmacy, GolpuraBarwala, Panchkula, Haryana, 134118, India
| | - Manni Rohilla
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Swami Vivekanand College of Pharmacy, Ram Nagar, Banur, Punjab, 140601, India
| | - Monika Saini
- Swami Vivekanand College of Pharmacy, Ram Nagar, Banur, Punjab, 140601, India
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133206, India
| | - Thakur Gurjeet Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Samrat Chauhan
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Soad K. Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Naseem Begum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 62529, Saudi Arabia
| | - Aziza Alshahrani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Mohammad Yousuf Ansari
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133206, India
| |
Collapse
|
11
|
Li Y, Wang GQ, Li YB. Therapeutic potential of natural coumarins in autoimmune diseases with underlying mechanisms. Front Immunol 2024; 15:1432846. [PMID: 39544933 PMCID: PMC11560467 DOI: 10.3389/fimmu.2024.1432846] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/14/2024] [Indexed: 11/17/2024] Open
Abstract
Autoimmune diseases encompass a wide range of disorders characterized by disturbed immunoregulation leading to the development of specific autoantibodies, which cause inflammation and multiple organ involvement. However, its pathogenesis remains unelucidated. Furthermore, the cumulative medical and economic burden of autoimmune diseases is on the rise, making these diseases a ubiquitous global phenomenon that is predicted to further increase in the coming decades. Coumarins, a class of aromatic natural products with benzene and alpha-pyrone as their basic structures, has good therapeutic effects on autoimmune diseases. In this review, we systematically highlighted the latest evidence on coumarins and autoimmune diseases data from clinical and animal studies. Coumarin acts on immune cells and cytokines and plays a role in the treatment of autoimmune diseases by regulating NF-κB, Keap1/Nrf2, MAPKs, JAK/STAT, Wnt/β-catenin, PI3K/AKT, Notch and TGF-β/Smad signaling pathways. This systematic review will provide insight into the interaction of coumarin and autoimmune diseases, and will lay a groundwork for the development of new drugs for autoimmune diseases.
Collapse
Affiliation(s)
- Yan Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Shandong Provincial Key Medical and Health Laboratory of Neuroimmunology, Jinan, China
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guan-qing Wang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Shandong Provincial Key Medical and Health Laboratory of Neuroimmunology, Jinan, China
| | - Yan-bin Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Shandong Provincial Key Medical and Health Laboratory of Neuroimmunology, Jinan, China
| |
Collapse
|
12
|
Shi G, Tai T, Miao Y, Yan L, Han T, Dong H, Liu Z, Cheng T, Liu Y, Yang Y, Fei S, Pang B, Chen T. The antagonism mechanism of astilbin against cadmium-induced injury in chicken lungs via Treg/Th1 balance signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116364. [PMID: 38657461 DOI: 10.1016/j.ecoenv.2024.116364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
The purpose of this study was to investigate the effect of Treg/Th1 imbalance in cadmium-induced lung injury and the potential protective effect of astilbin against cadmium-induced lung injury in chicken. Cadmium exposure significantly decreased T-AOC and GSH-Px levels and SOD activity in the chicken lung tissues. In contrast, it significantly increased the MDA and NO levels. These results indicate that cadmium triggers oxidative stress in lungs. Histopathological analysis revealed that cadmium exposure further induced infiltration of lymphocytes in the chicken lungs, indicating that cadmium causes pulmonary damage. Further analysis revealed that cadmium decreased the expression of IL-4 and IL-10 but increased those of IL-17, Foxp3, TNF-α, and TGF-β, indicating that the exposure of cadmium induced the imbalance of Treg/Th1. Moreover, cadmium adversely affected chicken lung function by activating the NF-kB pathway and inducing expression of genes downstream to these pathways (COX-2, iNOS), associated with inflammatory injury in the lung tissue. Astilbin reduced cadmium-induced oxidative stress and inflammation in the lungs by increasing antioxidant enzyme activities and restoring Treg/Th1 balance. In conclusion, our results suggest that astilbin treatment alleviated the effects of cadmium-mediated lung injury in chickens by restoring the Treg/Th1 balance.
Collapse
Affiliation(s)
- Guangliang Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Tiange Tai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Yusong Miao
- Heilongjiang Academy of Agricultural Sciences, Harbin, 150086, China
| | - Liangchun Yan
- Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China; Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu 610041, China
| | - Tianyu Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Han Dong
- Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China; Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu 610041, China
| | - Zhaoyang Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Tingting Cheng
- Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China; Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu 610041, China
| | - Yiding Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Yu Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Shanshan Fei
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Bo Pang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Tiezhu Chen
- Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China; Sichuan Provincial Key Laboratory of Quality and Innovation Research of Chinese Materia Medica, Chengdu 610041, China.
| |
Collapse
|
13
|
Pant T, Uche N, Juric M, Zielonka J, Bai X. Regulation of immunomodulatory networks by Nrf2-activation in immune cells: Redox control and therapeutic potential in inflammatory diseases. Redox Biol 2024; 70:103077. [PMID: 38359749 PMCID: PMC10877431 DOI: 10.1016/j.redox.2024.103077] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024] Open
Abstract
Inflammatory diseases present a serious health challenge due to their widespread prevalence and the severe impact on patients' lives. In the quest to alleviate the burden of these diseases, nuclear factor erythroid 2-related factor 2 (Nrf2) has emerged as a pivotal player. As a transcription factor intimately involved in cellular defense against metabolic and oxidative stress, Nrf2's role in modulating the inflammatory responses of immune cells has garnered significant attention. Recent findings suggest that Nrf2's ability to alter the redox status of cells underlies its regulatory effects on immune responses. Our review delves into preclinical and clinical evidence that underscores the complex influence of Nrf2 activators on immune cell phenotypes, particularly in the inflammatory milieu. By offering a detailed analysis of Nrf2's role in different immune cell populations, we cast light on the potential of Nrf2 activators in shaping the immune response towards a more regulated state, mitigating the adverse effects of inflammation through modeling redox status of immune cells. Furthermore, we explore the innovative use of nanoencapsulation techniques that enhance the delivery and efficacy of Nrf2 activators, potentially advancing the treatment strategies for inflammatory ailments. We hope this review will stimulate the development and expansion of Nrf2-targeted treatments that could substantially improve outcomes for patients suffering from a broad range of inflammatory diseases.
Collapse
Affiliation(s)
- Tarun Pant
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA; Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Nnamdi Uche
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matea Juric
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
14
|
Zhang Y, Xiao B, Liu Y, Wu S, Xiang Q, Xiao Y, Zhao J, Yuan R, Xie K, Li L. Roles of PPAR activation in cancer therapeutic resistance: Implications for combination therapy and drug development. Eur J Pharmacol 2024; 964:176304. [PMID: 38142851 DOI: 10.1016/j.ejphar.2023.176304] [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: 09/17/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 12/26/2023]
Abstract
Therapeutic resistance is a major obstacle to successful treatment or effective containment of cancer. Peroxisome proliferator-activated receptors (PPARs) play an essential role in regulating energy homeostasis and determining cell fate. Despite of the pleiotropic roles of PPARs in cancer, numerous studies have suggested their intricate relationship with therapeutic resistance in cancer. In this review, we provided an overview of the roles of excessively activated PPARs in promoting resistance to modern anti-cancer treatments, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy. The mechanisms through which activated PPARs contribute to therapeutic resistance in most cases include metabolic reprogramming, anti-oxidant defense, anti-apoptosis signaling, proliferation-promoting pathways, and induction of an immunosuppressive tumor microenvironment. In addition, we discussed the mechanisms through which activated PPARs lead to multidrug resistance in cancer, including drug efflux, epithelial-to-mesenchymal transition, and acquisition and maintenance of the cancer stem cell phenotype. Preliminary studies investigating the effect of combination therapies with PPAR antagonists have suggested the potential of these antagonists in reversing resistance and facilitating sustained cancer management. These findings will provide a valuable reference for further research on and clinical translation of PPAR-targeting treatment strategies.
Collapse
Affiliation(s)
- Yanxia Zhang
- School of Medicine, The South China University of Technology, Guangzhou, 510006, China; Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Bin Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yunduo Liu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Shunhong Wu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Qin Xiang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yuhan Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Junxiu Zhao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Ruanfei Yuan
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Keping Xie
- School of Medicine, The South China University of Technology, Guangzhou, 510006, China.
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| |
Collapse
|
15
|
Riaz F, Wei P, Pan F. PPARs at the crossroads of T cell differentiation and type 1 diabetes. Front Immunol 2023; 14:1292238. [PMID: 37928539 PMCID: PMC10623333 DOI: 10.3389/fimmu.2023.1292238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 11/07/2023] Open
Abstract
T-cell-mediated autoimmune type 1 diabetes (T1D) is characterized by the immune-mediated destruction of pancreatic beta cells (β-cells). The increasing prevalence of T1D poses significant challenges to the healthcare system, particularly in countries with struggling economies. This review paper highlights the multifaceted roles of Peroxisome Proliferator-Activated Receptors (PPARs) in the context of T1D, shedding light on their potential as regulators of immune responses and β-cell biology. Recent research has elucidated the intricate interplay between CD4+ T cell subsets, such as Tregs and Th17, in developing autoimmune diseases like T1D. Th17 cells drive inflammation, while Tregs exert immunosuppressive functions, highlighting the delicate balance crucial for immune homeostasis. Immunotherapy has shown promise in reinstating self-tolerance and restricting the destruction of autoimmune responses, but further investigations are required to refine these therapeutic strategies. Intriguingly, PPARs, initially recognized for their role in lipid metabolism, have emerged as potent modulators of inflammation in autoimmune diseases, particularly in T1D. Although evidence suggests that PPARs affect the β-cell function, their influence on T-cell responses and their potential impact on T1D remains largely unexplored. It was noted that PPARα is involved in restricting the transcription of IL17A and enhancing the expression of Foxp3 by minimizing its proteasomal degradation. Thus, antagonizing PPARs may exert beneficial effects in regulating the differentiation of CD4+ T cells and preventing T1D. Therefore, this review advocates for comprehensive investigations to delineate the precise roles of PPARs in T1D pathogenesis, offering innovative therapeutic avenues that target both the immune system and pancreatic function. This review paper seeks to bridge the knowledge gap between PPARs, immune responses, and T1D, providing insights that may revolutionize the treatment landscape for this autoimmune disorder. Moreover, further studies involving PPAR agonists in non-obese diabetic (NOD) mice hold promise for developing novel T1D therapies.
Collapse
Affiliation(s)
- Farooq Riaz
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Ping Wei
- Department of Otolaryngology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Fan Pan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
16
|
Liu P, Zhu W, Wang Y, Ma G, Zhao H, Li P. Chinese herbal medicine and its active compounds in attenuating renal injury via regulating autophagy in diabetic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1142805. [PMID: 36942026 PMCID: PMC10023817 DOI: 10.3389/fendo.2023.1142805] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Diabetic kidney disease (DKD) is the main cause of end-stage renal disease worldwide, and there is a lack of effective treatment strategies. Autophagy is a highly conserved lysosomal degradation process that maintains homeostasis and energy balance by removing protein aggregates and damaged organelles. Increasing evidence suggests that dysregulated autophagy may contribute to glomerular and tubulointerstitial lesions in the kidney under diabetic conditions. Emerging studies have shown that Chinese herbal medicine and its active compounds may ameliorate diabetic kidney injury by regulating autophagy. In this review, we summarize that dysregulation or insufficiency of autophagy in renal cells, including podocytes, glomerular mesangial cells, and proximal tubular epithelial cells, is a key mechanism for the development of DKD, and focus on the protective effects of Chinese herbal medicine and its active compounds. Moreover, we systematically reviewed the mechanism of autophagy in DKD regulated by Chinese herb compound preparations, single herb and active compounds, so as to provide new drug candidates for clinical treatment of DKD. Finally, we also reviewed the candidate targets of Chinese herbal medicine regulating autophagy for DKD. Therefore, further research on Chinese herbal medicine with autophagy regulation and their targets is of great significance for the realization of new targeted therapies for DKD.
Collapse
Affiliation(s)
- Peng Liu
- Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| | - Wenhui Zhu
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Yang Wang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Guijie Ma
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Hailing Zhao
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Hailing Zhao, ; Ping Li,
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Hailing Zhao, ; Ping Li,
| |
Collapse
|