1
|
Mavaddatiyan L, Naeini S, Khodabandeh S, Hosseini F, Skelton RP, Azizi V, Talkhabi M. Exploring the association between aging, ferroptosis, and common age-related diseases. Arch Gerontol Geriatr 2025; 135:105877. [PMID: 40339241 DOI: 10.1016/j.archger.2025.105877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 04/14/2025] [Accepted: 04/26/2025] [Indexed: 05/10/2025]
Abstract
Aging is a natural biological process that is characterized by the progressive decline in physiological functions and an increased vulnerability to age-related diseases. The aging process is driven by different cell and molecular mechanisms. It has recently been shown that aging is associated with heightened vulnerability to ferroptosis (an intracellular iron-dependent form of programmed cell death). This susceptibility arises from various factors including oxidative stress, impaired antioxidant defences, and dysregulated iron homeostasis. The progressive decline in cellular antioxidant capacity and the accumulation of damaged components contribute to the increased susceptibility of aging cells to ferroptosis. Dysregulation of key regulators involved in ferroptosis, such as glutathione peroxidase 4 (GPX4), iron regulatory proteins, and lipid metabolism enzymes, further exacerbates this vulnerability. The decline in cellular defence mechanisms against ferroptosis during aging contributes to the accumulation of damaged cells and tissues, ultimately resulting in the manifestation of age-related diseases. Understanding the intricate relevance between aging and ferroptosis holds significant potential for developing strategies to counteract the detrimental effects of aging and age-related diseases. This will subsequently act to mitigate the negative consequences of aging and improving overall health in the elderly population. This review aims to clarify the relationship between aging and ferroptosis, and explores the underlying mechanisms and implications for age-related disorders, including neurodegenerative, cardiovascular, and neoplastic diseases. We also discuss the accumulating evidence suggesting that the imbalance of redox homeostasis and perturbations in iron metabolism contribute to the age-associated vulnerability to ferroptosis.
Collapse
Affiliation(s)
- Laleh Mavaddatiyan
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - SaghiHakimi Naeini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Sara Khodabandeh
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Fatemeh Hosseini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - RhysJ P Skelton
- Flinders Medical Centre, Department of Ophthalmology, Bedford Park, Australia
| | - Vahid Azizi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mahmood Talkhabi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
2
|
Li J, Gu J, Pan S, Deng N, Khan M, Li L, Wu X, Li Y. Synergic effect of the combination of isoliquiritigenin and arsenic trioxide in HepG2 liver cancer cells. Cell Signal 2025; 131:111752. [PMID: 40107478 DOI: 10.1016/j.cellsig.2025.111752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/06/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
Despite continuous therapeutic interventions, the prognosis of hepatocellular carcinoma (HCC) remains very poor. Thus, quest for novel treatment strategies to improve therapeutic window of HCC therapy is paramount. Arsenic trioxide (ATO) is commonly used as the first-line treatment for acute promyelocytic leukemia (APL). Isoliquiritigenin (ISL) is a potential plant-based bioactive molecule with versatile biological and pharmacological effects including anticancer effect. The present study aimed to investigate the potential synergistic effects of combination of ISL and ATO in HCC cells. The data revealed that the combination of ISL and ATO synergistically inhibited HCC cell proliferation. The collective data demonstrate that synergistic anticancer effect of combined treatment of ISL + ATO was achieved via cooperative induction of mitochondrial apoptosis through ROS generation and inhibition of PI3K/Akt/mTOR pathway. In addition, ROS generation and suppression of PI3K/Akt/mTOR pathway were found to be two independent events in induction of apoptosis. Finally, we observed that combination treatment effectively suppressed tumor growth in nude mice xenograft model through induction of intrinsic apoptosis and inhibition of PI3K/Akt/mTOR pathway. In conclusion, the findings of this study suggest that both drugs work synergistically to exert anti-tumor effect in HCC, both in-vitro and in-vivo and could offer novel strategy for liver cancer treatment.
Collapse
Affiliation(s)
- Jingjing Li
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Juan Gu
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sijia Pan
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Nuo Deng
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Muhammad Khan
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan.
| | - Lingyan Li
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiao Wu
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yongming Li
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Han D, Yu Z, Zhang K, Gai C, Zhang P, Chai X, Zhuo X, Zhao Q, Zou Y, Zhu L. Design, synthesis, and antitumor activity of stapled peptide inhibitors targeting the RAS-RAF interactions. Eur J Med Chem 2025; 290:117568. [PMID: 40153928 DOI: 10.1016/j.ejmech.2025.117568] [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: 01/03/2025] [Revised: 03/12/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
RAS-RAF interactions play a vital role in the RAS-RAF-MEK-ERK signaling pathway, significantly regulating cell proliferation, differentiation, and survival. Some small molecule inhibitors targeting various components of this pathway, such as MRTX849 and AMG 510, have been introduced for clinical application. However, peptide-based drugs encounter several challenges, such as poor cell permeability, low biological stability, and rapid in vivo clearance, which hinder their application. Herein, based on co-crystal complex structures and RAS-RAF interaction hotspots, we identified four linear peptides-Raf-0 to Raf-2 and CRD-0-derived from the α-helical regions of the RAS-binding domain (RBD) and the cysteine-rich domain (CRD) of CRAF. Raf-1 was selected for further modification using a hydrocarbon stapling strategy, capping it with stearic acid at the N-terminal due to its highest binding affinity in the SPR assay. As a result, Sraf-2-1 and Sraf-7-1 bound to KRASG12C with Kd values of 3.56 μM and 2.62 μM, respectively, demonstrating robust anticancer activity in the CCK8 assay. Additionally, Sraf-2-1 and Sraf-7-1 reduced AKT phosphorylation, induced cancer cell apoptosis in a concentration-dependent manner, and effectively inhibited cancer cell migration, showing improved α-helix stability and cell permeability. In summary, our findings indicate that the hydrocarbon stapling strategy and stearic acid tagging enhanced the therapeutic potential of peptide inhibitors, offering methods for targeting RAS in cancer therapy.
Collapse
Affiliation(s)
- Dan Han
- School of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, PR China; Department of Burn Plastic Surgery, The Second Affiliated Hospital of Second Military Medical University, Shanghai, 200003, PR China
| | - Zhou Yu
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Kai Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Conghao Gai
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Peichao Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Xiaoyun Chai
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Xiaobing Zhuo
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Qingjie Zhao
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China.
| | - Yan Zou
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China.
| | - Lie Zhu
- School of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, PR China; Department of Burn Plastic Surgery, The Second Affiliated Hospital of Second Military Medical University, Shanghai, 200003, PR China.
| |
Collapse
|
4
|
Zhu X, Miao L, Li J, Wang X, Lin L, Zhang Y, Sun M, Meng S, Ren J, Peng Q, Liu J. Exploring the effective components and underlying mechanisms of Feiyanning formula in acute lung injury based on the pharmacokinetics, metabolomics and network pharmacology technology. Fitoterapia 2025; 183:106486. [PMID: 40120984 DOI: 10.1016/j.fitote.2025.106486] [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: 11/27/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
This study aimed to explore the mechanisms of Feiyanning formula (FYN) on acute lung injury (ALI) using pharmacokinetics combined with network pharmacology strategy. Firstly, pharmacokinetic studies of 13 major bioactive components in normal and ALI mice were conducted using ultra-high performance liquid chromatography-triple quadrupole mass spectrometry (UPLC-QQQ-MS/MS). Secondly, metabolomics was utilized to explore the metabolites affected by FYN. Finally, the network pharmacology was used to analyze the pharmacological mechanism of FYN's pharmacokinetic target components in ALI treatment, with western blotting (WB) experiment performed for verification. The pharmacokinetic results showed that compared to normal mice, the Cmax and AUC0-t of wogonin, oroxylin A, liquiritigenin, tetrandrine, and fangchinoline were significantly increased in ALI mice. The results of the lung tissue distribution showed that compared to normal mice, the AUC0-t of wogonin and oroxyloside was significantly increased in ALI mice; the Cmax of wogonoside and norwogonin was significantly increased in ALI mice. Metabolomics analysis showed that FYN may alleviate LPS-induced lung inflammation in mice by regulating related pathways including purine metabolism, and phenylalanine, tyrosine and tryptophan biosynthesis in both serum and lung tissue. Network pharmacology identified 110 overlapping genes between the 13 absorbed components and ALI-related targets. In KEGG enrichment analysis, the PI3K/AKT signaling pathway was identified as a significant pathway. WB experiment confirmed that FYN reduced the expression ratios of p-PI3K/PI3K, p-AKT1/AKT1, p-EGFR/EGFR, and TLR4 levels in lung tissue of ALI mice. This study might offer a solid foundation for evaluating the clinical efficacy of FYN.
Collapse
Affiliation(s)
- Xuanxuan Zhu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China; School of Traditional Chinese Pharmacy, Bozhou University, Bozhou, China
| | - Lan Miao
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China
| | - Junmei Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China
| | - Xinwei Wang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China; Research Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Lin
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China
| | - Ying Zhang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China
| | - Mingqian Sun
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China
| | - Shuo Meng
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China
| | - Junguo Ren
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China
| | - Qing Peng
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China.
| | - Jianxun Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Pharmacology of Traditional Chinese Medicine, Beijing, China.
| |
Collapse
|
5
|
Dan LX, Xie SP. Autophagy in cardiac pathophysiology: Navigating the complex roles and therapeutic potential in cardiac fibrosis. Life Sci 2025:123761. [PMID: 40419108 DOI: 10.1016/j.lfs.2025.123761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 05/07/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
Cardiac fibrosis is a critical factor in cardiac structural remodeling and dysfunction, closely associated with the progression of various cardiovascular diseases (CVDs), including heart failure and myocardial infarction (MI). It is characterized by excessive extracellular matrix (ECM) deposition, which disrupts normal cardiac architecture and impairs cardiac function. Autophagy, a cellular degradation and recycling mechanism, is essential for maintaining cardiac homeostasis, mitigating stress responses, and preventing cellular damage. Recent studies have revealed a significant link between autophagy and cardiac fibrosis, suggesting that autophagic dysregulation can exacerbate fibrosis by promoting fibroblast activation and ECM accumulation. Conversely, proper autophagic activity may attenuate cardiac fibrosis by removing damaged cellular components and regulating fibrotic signaling pathways. This review examines the role of autophagy in cardiac fibrosis. It also emphasizes potential pharmacological strategies that can be used to modulate autophagic processes. These strategies may serve as therapeutic approaches for treating cardiac fibrosis, with the ultimate goal of preventing excessive fibrosis and enhancing cardiac function.
Collapse
Affiliation(s)
- Ling-Xuan Dan
- Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Song-Ping Xie
- Renmin Hospital of Wuhan University, Wuhan 430060, PR China.
| |
Collapse
|
6
|
Liu C, Wang X, Xu S, Liu M, Cao X. Regulation of autophagy: Insights into O-GlcNAc modification mechanisms. Life Sci 2025; 369:123547. [PMID: 40058573 DOI: 10.1016/j.lfs.2025.123547] [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: 12/28/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/30/2025]
Abstract
Autophagy is a "self-eating" biological process that degrades cytoplasmic contents to ensure cellular homeostasis. Its response to stimuli occurs in two stages: Within a few to several hours of exposure to a stress condition, autophagic flow rapidly increases, which is mediated by post-translational modification (PTM). Subsequently, the transcriptional program is activated and mediates the persistent autophagic response. O-linked β-N-acetylglucosamine (O-GlcNAc) modification is an inducible and dynamically cycling PTM; mounting evidence suggests that O-GlcNAc modification participates in the total autophagic process, including autophagy initiation, autophagosome formation, autophagosome-lysosome fusion, and transcriptional process. In this review, we summarize the current knowledge on the emerging role of O-GlcNAc modification in regulating autophagy-associated proteins and explain the different regulatory effects on autophagy exerted by O-GlcNAc modification.
Collapse
Affiliation(s)
- Chengzhi Liu
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinyu Wang
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Shengnan Xu
- College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Mingyue Liu
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xusheng Cao
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China.
| |
Collapse
|
7
|
Liu L, Wuyun T, Sun X, Zhang Y, Cha G, Zhao L. Therapeutic efficacy of TMTP1-modified EVs in overcoming bone metastasis and immune resistance in PIK3CA mutant NSCLC. Cell Death Dis 2025; 16:367. [PMID: 40328748 PMCID: PMC12055990 DOI: 10.1038/s41419-025-07685-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025]
Abstract
Non-small cell lung cancer (NSCLC) with PIK3CA mutations demonstrates significant challenges in treatment due to enhanced bone metastasis and immune checkpoint resistance. This study investigates the efficacy of tumor-targeting peptide 1-modified cancer stem cell-derived extracellular vesicles (TMTP1-TSRP-EVs) in reshaping the tumor microenvironment and reversing immune checkpoint resistance in NSCLC. By integrating TMTP1-TSRP into EVs, we aim to specifically deliver therapeutic agents to NSCLC cells, focusing on inhibiting the PI3K/Akt/mTOR pathway, a crucial driver of oncogenic activity and immune evasion in PIK3CA-mutated cells. Our comprehensive in vitro and in vivo analyses show that TMTP1-TSRP-EVs significantly inhibit tumor growth, reduce PD-L1 expression, and enhance CD8+ T cell infiltration, effectively reversing the immune-suppressive microenvironment. Moreover, the in vivo models confirm that our approach not only suppresses bone metastases but also overcomes primary resistance to immune checkpoint inhibitors by modulating the expression of key immunological markers. These findings suggest that targeted delivery of TMTP1-TSRP-EVs could provide a novel therapeutic strategy for treating PIK3CA-mutant NSCLC, offering significant improvements over traditional therapies by directly targeting the molecular pathogenesis of tumor resistance and metastasis. Molecular Mechanisms Reshaping the TME to Halt PI3K-Mutant Bone Metastasis of NSCLC and Overcoming Primary ICI Resistance. (Created by BioRender).
Collapse
Affiliation(s)
- Liwen Liu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tanghesi Wuyun
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Sun
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Zhang
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Geqi Cha
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ling Zhao
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
8
|
Moreno-Estar S, Cidad P, Arevalo-Martinez M, Portillo AM, Sacristan-Moraleda M, Alonso E, Lopez-Lopez JR, Perez-Garcia MT. Vascular Smooth Muscle Cell Migration and P70S6K: Key Players in Intimal Hyperplasia Development. J Am Heart Assoc 2025; 14:e038358. [PMID: 40314369 DOI: 10.1161/jaha.124.038358] [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: 08/15/2024] [Accepted: 03/19/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Vascular smooth muscle cell (VSMC) recruitment and activation by vessel injury cause intimal hyperplasia (IH) and restenosis. Drug-eluting stents releasing mTOR (mechanistic target of rapamycin) blockers (sirolimus, everolimus [EV]) improve surgery outcomes but exhibit nonspecific effects and poor efficacy in diseased vessels. Drug combinations targeting the multifactorial processes leading to IH could enhance efficacy and reduce toxicity. Our previous work showed that Kv1.3 channel blockers such as 5-(4-phenoxybutoxy)psoralen (PAP-1) prevented IH. Since Kv1.3 signaling works through the MEK/ERK pathway, we hypothesize that PAP-1 and EV combination could improve antirestenotic therapies. METHODS AND RESULTS The effects of PAP-1, EV, and their combination on IH development were studied in vivo using a carotid ligation mouse model and ex vivo in organ culture of human vessels. Individually, both drugs inhibited vessel remodeling, but, surprisingly, their combination canceled these inhibitory effects. In primary human VSMCs cultures, the drug combination abolished the inhibition of cell migration but not cell proliferation, which was even potentiated. We uncovered a crosstalk between mTOR and MEK/ERK pathways in VSMCs, centered on P70S6K activation. P70S6K phosphorylation levels correlated with IH development, even reproducing the differences in EV response between diabetic and nondiabetic samples. CONCLUSIONS VSMC migration, rather than proliferation, mirrors PAP-1 and EV effects on IH development in vessels. Critically, we identify VSMC P70S6K phosphorylation as a surrogate marker for IH progression. The nonmonotonic responses of P70S6K activation to pathway blockers suggest the existence of a crosstalk element functioning as an exclusive NOR logic gate providing new insights for IH prevention strategies.
Collapse
MESH Headings
- Animals
- Ribosomal Protein S6 Kinases, 70-kDa/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Cell Movement/drug effects
- Hyperplasia
- Humans
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Everolimus/pharmacology
- Disease Models, Animal
- Neointima
- Mice, Inbred C57BL
- Cell Proliferation/drug effects
- Cells, Cultured
- Mice
- Male
- TOR Serine-Threonine Kinases/metabolism
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- Signal Transduction
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/enzymology
- Phosphorylation
- Vascular Remodeling/drug effects
- MTOR Inhibitors/pharmacology
Collapse
Affiliation(s)
- Sara Moreno-Estar
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| | - Pilar Cidad
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| | - Marycarmen Arevalo-Martinez
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| | - Ana M Portillo
- Departamento de Matemática Aplicada Universidad de Valladolid Instituto de Investigación en Matemáticas Valladolid Spain
| | - Marcos Sacristan-Moraleda
- Departamento de Matemática Aplicada Universidad de Valladolid Instituto de Investigación en Matemáticas Valladolid Spain
| | - Esperanza Alonso
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| | - Jose R Lopez-Lopez
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| | - M Teresa Perez-Garcia
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| |
Collapse
|
9
|
Yu B, Cao Y, Lin P, Zhang L, Chen M. Enhancement of Ndrg2 promotes hypertrophic scar fibrosis by regulating PI3K/AKT signaling pathway. Cell Signal 2025; 129:111659. [PMID: 39956247 DOI: 10.1016/j.cellsig.2025.111659] [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: 11/15/2024] [Revised: 01/17/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Hypertrophic scar (HTS) is a prevalent chronic inflammatory skin disorder characterized by abnormal proliferation and extracellular matrix deposition. N-Myc downstream regulated gene 2 (Ndrg2) is a cell stress response gene related to cell proliferation, differentiation and various fibrotic diseases. However, the role of Ndrg2 in HTS is unknown and warrants further investigation. In this study, we confirmed that the expression of Ndrg2 was increased in HTS of human and a bleomycin-induced fibrosis mouse model. We then used Ndrg2 knockout mice and found Ndrg2 deletion could significantly reduce the synthesis of collagen and alleviate skin fibrosis. In addition, the proliferation and migration of Ndrg2-interfered HTS-derived fibroblasts decreased and those of Ndrg2-overexpressed normal skin-derived fibroblasts increased. Further, by western blot analysis, we verified that the expression of phosphorylated-PI3K, PI3K, phosphorylated-AKT and AKT were all increased after Ndrg2 overexpressed in normal skin-derived fibroblasts. Moreover, PI3K inhibitor (LY294002) administration significantly rescued the effect of Ndrg2 overexpression on skin fibrosis. In summary, our results demonstrated that Ndrg2 could promote HTS fibrosis by mediating PI3K/AKT signaling pathway. Our data suggest that Ndrg2 may be a promising therapeutic target for HTS.
Collapse
Affiliation(s)
- Boya Yu
- Department of Plastic and Reconstructive Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; Chinese PLA Medical School, Beijing 100853, China.
| | - Yalei Cao
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Pianpian Lin
- Department of Plastic and Reconstructive Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; Chinese PLA Medical School, Beijing 100853, China
| | - Lixia Zhang
- Department of Plastic and Reconstructive Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; Chinese PLA Medical School, Beijing 100853, China.
| | - Minliang Chen
- Department of Plastic and Reconstructive Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; Chinese PLA Medical School, Beijing 100853, China.
| |
Collapse
|
10
|
Guo S, Ding R, Zhao Q, Wang X, Lv S, Ji XY. Recent Insights into the Roles of PEST-Containing Nuclear Protein. Mol Biotechnol 2025; 67:1800-1813. [PMID: 38762838 DOI: 10.1007/s12033-024-01188-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 04/26/2024] [Indexed: 05/20/2024]
Abstract
PEST-containing nuclear protein (PCNP), a short-lived small nuclear protein with 178 amino acids, is a nuclear protein containing two PEST sequences. PCNP is highly expressed in several malignant tumors such as cervical cancer, rectal cancer, and lung cancer. It is also associated with cell cycle regulation and the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and Wnt signaling pathways during tumor growth. The present article discuss how PCNP regulates the PI3K/AKT/mTOR and Wnt signaling pathways and related proteins, and the ubiquitination of PCNP regulates tumor cell cycle as well as the progress of the application of PCNP in the pathophysiology and treatment of colon cancer, human ovarian cancer, thyroid cancer, lung adenocarcinoma and oral squamous cell carcinoma. The main relevant articles were retrieved from PubMed, with keywords such as PEST-containing nuclear protein (PCNP), cancer (tumor), and signaling pathways as inclusion/exclusion criteria. Relevant references has been included and cited in the manuscript.
Collapse
Affiliation(s)
- Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Ruidong Ding
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Qian Zhao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Xu Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Shuangyu Lv
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
- Kaifeng Key Laboratory for Infectious Diseases and Biosafety, Kaifeng, 475004, Henan, China.
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Mazhai, Erqi District, Zhengzhou, 450064, Henan, China.
| |
Collapse
|
11
|
Zhang Y, Tan BY, Peng HZ, Duan Y, Wen X, Li XY, Wu XR, Zhao YL, Luo XD. 10-Methoxy-leonurine accelerated wound healing through ErbB4/PI3K-AKT pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119641. [PMID: 40118194 DOI: 10.1016/j.jep.2025.119641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Leonurusjaponicus Houtt., commonly known as motherwort, has been used in traditional Chinese medicine to treat trauma and wound infections historically. However, the main compounds responsible for promoting wound healing need to be identified and explained fully. AIM OF THE STUDY To investigate the wound healing effect of 10-methoxy-leonurine (MN) and explore its mechanism. MATERIALS AND METHODS Following the UHPLC/Q-TOF-MS analysis of L. japonicus, the phytochemical isolation was carried out, and then the effect of isolated compounds in promoting the proliferation of human skin fibroblasts (HSF) was screened. Additionally, scratch and 5-ethynyl-2'-deoxyuridine assay in HSF were further adopted to support the effects of MN in vitro. A rat model with full-thickness skin wounds was used to verify in vivo by hematoxylin and eosin staining, Masson's trichrome and immunohistochemistry. Furthermore, network pharmacology and molecular docking were performed to predict the potential pathway, which were subsequently validated by cell cycle and RT-qPCR. RESULTS A total 11 compounds were isolated, in which MN exhibited the most significant bioactivity in promoting HSF proliferation and migration. Moreover, the wound healing rates in low (10 μg/mL) and high dose (50 μg/mL) of MN groups reached 93.9 % and 94.5 %, respectively, which was better than basic fibroblast growth factor (bFGF) 89.4 %. Additionally, the epithelial thickness, collagen Ⅰ deposition and α-smooth muscle actin expression were enhanced and the proportion of HSF cells in the S phase of the cell cycle was increased. Network pharmacological and molecular docking analysis suggested ErbB pathway was involved in the regulation of wound healing by MN, which was further supported by the up-regulation of Erbb4, Stat5, Pi3k, Akt and mTOR mRNA levels. CONCLUSION MN showed potent effect on wound healing by regulating ErbB4/PI3K-AKT pathway, even better than bFGF.
Collapse
Affiliation(s)
- Yue Zhang
- Yunnan Characteristic Plant Extraction Laboratory Co. Ltd, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, PR China
| | - Bang-Yin Tan
- Yunnan Characteristic Plant Extraction Laboratory Co. Ltd, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, PR China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, PR China
| | - Hui-Zhen Peng
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, School of Basic Medicine, Yunnan University of Chinese Medicine, Southwest United Graduate School, Kunming, 650500, PR China
| | - Yu Duan
- Yunnan Characteristic Plant Extraction Laboratory Co. Ltd, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, PR China
| | - Xi Wen
- Key Laboratory of Natural Drug Pharmacology, School of Pharmacy, Kunming Medical University, Kunming, 650500, PR China
| | - Xin-Yao Li
- Yunnan Characteristic Plant Extraction Laboratory Co. Ltd, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, PR China
| | - Xian-Run Wu
- Yunnan Characteristic Plant Extraction Laboratory Co. Ltd, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, PR China
| | - Yun-Li Zhao
- Yunnan Characteristic Plant Extraction Laboratory Co. Ltd, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, PR China.
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory Co. Ltd, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, PR China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, PR China.
| |
Collapse
|
12
|
Yang Y, Wu G, Wang Y, Mao Q, Zhang D, Wu J. LL37 promotes angiogenesis: a potential therapeutic strategy for lower limb ischemic diseases. Front Pharmacol 2025; 16:1587351. [PMID: 40337519 PMCID: PMC12055537 DOI: 10.3389/fphar.2025.1587351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/08/2025] [Indexed: 05/09/2025] Open
Abstract
Purpose To study the angiogenic capacity of antimicrobial peptide LL37 (cathelicidin antimicrobial peptide), explore its molecular mechanisms, and provide new ideas for treating lower limb ischemic diseases. Methods LL37 was applied exogenously to human umbilical vein endothelial cells (HUVECs), and its effects on cell proliferation, migration, and angiogenesis were assessed using Cell Counting Kit-8 (CCK-8), plate cloning, scratch, and angiogenesis assays. A mouse lower limb ischemia model was established, with LL37 injected intramuscularly on days 0, 4, and 8. Blood flow recovery was evaluated by laser Doppler flowmetry. Immunofluorescence staining detected cluster of differentiation 31 (CD31) and cluster of differentiation 34 (CD34) expression, while Hematoxylin and Eosin (H&E) staining assessed muscle cell morphology. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting analyzed gene and protein expression changes in HUVECs. Results LL37 enhanced the proliferative, migratory, and pro-angiogenic abilities of HUVECs. It significantly improved blood flow recovery in ischemic limbs, with higher CD31/CD34 expression and more intact muscle morphology. qRT-PCR analysis demonstrated elevated expression of angiogenesis-related genes in LL37-treated HUVECs. Western blotting revealed increased vascular endothelial growth factor A (VEGFA) expression and enhanced phosphorylation levels of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway in LL37-treated cells. Conclusion LL37 promotes angiogenesis via the VEGFA-PI3K/AKT/mTOR pathway, showing potential for treating lower limb ischemia by improving perfusion.
Collapse
Affiliation(s)
| | | | | | | | | | - Jitao Wu
- *Correspondence: Dongxu Zhang, ; Jitao Wu,
| |
Collapse
|
13
|
Tan C, Cong S, Xie Y, Zhi Y. Mechanistic Integration of Network Pharmacology and In Vivo Validation: TFRD Combat Osteoporosis via PI3K/AKT Pathway Activation. Int J Mol Sci 2025; 26:3650. [PMID: 40332238 PMCID: PMC12027698 DOI: 10.3390/ijms26083650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
In the context of osteoporosis closely linked to bone metabolism imbalance caused by estrogen deficiency, total flavonoids of Rhizoma Drynariae (TFRD) exhibit potential anti-osteoporotic activity, yet their multicomponent synergistic mechanism and association with the PI3K/AKT signaling pathway remain unclear. This study aimed to systematically elucidate the molecular mechanisms by which TFRD regulate bone metabolism and improve osteoporosis in ovariectomized (OVX) rats through the PI3K/AKT pathway, integrating network pharmacological predictions with animal experimental validation. Methods involved identifying TFRD's active components using UPLC/MS-MS, predicting targets with SwissTargetPrediction, constructing a "component-target-disease" network, and performing GO/KEGG enrichment analysis with MetaScape (v3.5). In vivo experiments established an OVX rat model, randomized into sham, OVX, low-/high-dose TFRD, and sim groups, assessing bone mineral density (BMD) and mandibular Micro-CT parameters after 12 weeks. Western blot analyzed PI3K, p-AKT1, and related protein expressions. Results showed the high-dose TFRD group significantly increased BMD, improved trabecular bone quantity and structure, and upregulated PI3K, p-PI3K, and p-AKT1 protein expressions compared to the OVX group. Molecular docking confirmed stable binding energy between core components and PI3K/AKT targets. TFRD may ameliorate estrogen deficiency-induced osteoporosis by activating the PI3K/AKT signaling pathway, inhibiting bone resorption, and promoting osteogenic differentiation, providing pharmacological evidence for multitarget treatment of osteoporosis with traditional Chinese medicine.
Collapse
Affiliation(s)
- Chang Tan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| | - Shibo Cong
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| | - Yanming Xie
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| | - Yingjie Zhi
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| |
Collapse
|
14
|
Zhang J, Zhao L, He J, Wu H, Guo M, Yu Z, Ma X, Yong Y, Li Y, Ju X, Liu X. Protect Effects of Perilla Seed Extract and Its Active Ingredient Luteolin Against Inflammatory Bowel Disease Model via the PI3K/AKT Signal Pathway In Vivo and In Vitro. Int J Mol Sci 2025; 26:3564. [PMID: 40332054 PMCID: PMC12026851 DOI: 10.3390/ijms26083564] [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: 02/06/2025] [Revised: 03/19/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
The purpose of this study was to investigate the anti-inflammatory effects of Perilla Seed Extract (PSE) and its active ingredient on Inflammatory Bowel Disease (IBD) in vitro and in vivo. Thirty-two C57/BL mice were randomly divided into four groups (n = 8): control group (CON), PBS group, LPS group (LPS 3.5 mg/kg given intraperitoneally [ip] on day 7 of the study only), and PSE group (100 mg/kg orally daily + LPS ip at 3.5 mg/kg on day 7). Mice were euthanized 24 h after LPS administration. MODE-K cells were divided into five groups: control group (CON), LPS group (50 μg/mL LPS for 2 h), and PSE group (low dose, 25 μg/mL PSE + LPS; middle dose, 50 μg/mL PSE + LPS; high dose, 100 μg/mL PSE + LPS). In vivo, compared with the CON group, LPS revealed a significant decrease in the villus length-to-crypt depth ratio (p < 0.01) and goblet cell density per unit area (p < 0.01). Conversely, PSE administration resulted in a significant increase in the villus length-to-crypt depth ratio (p < 0.01) and goblet cell density (p < 0.01). LPS significantly increased the ROS content (p < 0.01), the secretion of inflammatory cytokines of IL-6 (p < 0.01), TNF-α (p < 0.01), and the mRNA expressions of HO-1 (p < 0.01). LPS significantly decreased the mRNA expressions of Occludin (p < 0.01) and Claudin1 (p < 0.01). In contrast, PSE treatment led to a marked decrease in ROS levels (p < 0.01), along with a reduction in the secretion of inflammatory factors IL-6 (p < 0.01) and TNF-α(p < 0.05), as well as the mRNA expressions of HO-1 (p < 0.01). Concurrently, PSE significantly increased the mRNA expressions of Occludin (p < 0.05) and Claudin1 (p < 0.01). In vitro, PSE treatment also significantly reversed LPS-induced inflammation, oxidation and tight junction-related factors. Network pharmacology identified 97 potential targets for PSE in treating IBD, while transcriptomics analysis revealed 342 differentially expressed genes (DEGs). Network pharmacology and transcriptomics analysis indicated that significant pathways included the PI3K-Akt signaling pathway, MAPK signaling pathway, and TNF signaling pathway, of which the PI3K-AKT pathway may represent the primary mechanism. In an in vivo setting, compared with the CON group, LPS led to a significant increase in the protein expression of p-PI3K/PI3K (p < 0.01) and p-AKT1/AKT1 (p < 0.01). Conversely, PSE resulted in a significant decrease in the protein expression of p-PI3K/PI3K (p < 0.01) and p-AKT1/AKT1 (p < 0.01). In vitro, compared with the LPS group, PSE also significantly blocked the protein expression of p-PI3K/PI3K (p < 0.01) and p-AKT1/AKT1 (p < 0.01). The chemical composition of PSE was analyzed using UPLC-MS/MS, which identified six components including luteolin (content 0.41%), rosmarinic acid (content 0.27%), α-linolenic acid (content 1.2%), and oleic acid (content 0.2%). Molecular docking found that luteolin could establish stable binding with eight targets, and luteolin significantly decreased the p-AKT1/AKT1 ratio (p < 0.01) compared to the LPS group in MODE-K cells. In summary, PSE demonstrates efficacy against IBD progression by enhancing intestinal barrier function and inhibiting inflammatory responses and oxidative stress via the PI3K/AKT signaling pathway, and luteolin's inhibition of AKT1 protein phosphorylation appears to play a particularly crucial role in this therapeutic mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xiaoxi Liu
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524091, China; (J.Z.)
| |
Collapse
|
15
|
Xu MZ, Ke F, Chai JP, A JD, Tan QL. Progress on the HIF-1α/VEGF/VEGFR2 signal pathway in hepatic alveolar echinococcosis. Front Oncol 2025; 15:1553125. [PMID: 40265025 PMCID: PMC12011584 DOI: 10.3389/fonc.2025.1553125] [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: 12/30/2024] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
Alveolar echinococcosis (AE), a lethal parasitic zoonosis mimicking malignant tumors, progresses via hepatic infiltration and metastatic spread, causing multiorgan failure. Despite its clinical resemblance to cancer, molecular drivers of its aggressiveness remain poorly defined. Recent studies highlight perilesional angiogenesis as pivotal for lesion invasiveness, mediated by VEGF-driven pathological vascularization. VEGF not only fuels parasitic proliferation by creating nutrient-rich microenvironments but also engages crosstalk with host-parasite interactions, including immune evasion by Echinococcus multilocularis, germinal layer hyperplasia, and periparasitic inflammation.Targeting the HIF-1α/VEGF/VEGFR2 axis emerges as a promising therapeutic strategy. Mechanistically, VEGF/VEGFR2 blockade may simultaneously disrupt angiogenesis-dependent parasitic expansion and survival pathways. Preclinical evidence shows that inhibiting HIF-1α (VEGF's upstream regulator) suppresses metacestode proliferation and tissue invasion by starving lesions of vascular support while modulating immune-inflammatory responses. This dual action addresses both parasitic resource acquisition and host defence subversion.This review synthesizes molecular insights into HIF-1α/VEGF-mediated pathogenesis with clinical observations, proposing anti-angiogenic therapy as a rational adjunct to current treatments. By delineating VEGF's role in sustaining parasitic metabolic demands and immune regulation, we underscore the translational potential of pathway-specific inhibitors. Such approaches could mitigate limitations of conventional therapies (e.g., benzimidazoles), particularly for advanced-stage AE with microvascular proliferation. Systematic analysis of angiogenesis signalling networks advances our understanding of AE's "parasitic cancer" paradigm while guiding development of targeted interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Meng-Zhao Xu
- The Graduate School, Qinghai University, Xining, China
| | - Fei Ke
- The Graduate School, Qinghai University, Xining, China
| | - Jin-Ping Chai
- Department of Internal Medicine-Cardiovascular, Qinghai Provincial People’s Hospital, Xining, China
| | - Ji-De A
- Department of Hepatic Hydatidosis, Qinghai Provincial People’s Hospital, Xining, China
| | - Qing-Long Tan
- Department of General Surgery, Qinghai Provincial People’s Hospital, Xining, China
| |
Collapse
|
16
|
Liu B, Yao Z, Song L, Sun C, Shen C, Cheng F, Cheng Z, Zhang R, Liu R. Vitexin alleviates lipid metabolism disorders and hepatic injury in obese mice through the PI3K/AKT/mTOR/SREBP-1c pathway. Eur J Med Chem 2025; 287:117379. [PMID: 39947052 DOI: 10.1016/j.ejmech.2025.117379] [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: 11/21/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/24/2025]
Abstract
Obesity is recognized as a metabolic disorder, and its treatment and management pose ongoing challenges worldwide. Hawthorn, a traditional Chinese herb used to alleviate digestive issues and reduce blood lipid levels, has unclear mechanisms of action regarding its active components in the treatment of obesity. This study investigated the anti-obesity effects of vitexin, a major flavonoid compound found in hawthorn, in high-fat diet (HFD)-induced C57BL/6 mice. The results demonstrated that vitexin significantly reduced body weight, liver weight, blood lipid levels, and inflammatory markers in obese mice, while also inhibiting hepatic lipid accumulation. Mechanistic studies revealed that vitexin likely suppresses adipogenesis by modulating the PI3K-AKT signaling pathway, as evidenced by reduced expression of PI3K, phosphorylated AKT, phosphorylated mTOR, and SREBP-1c in the livers of vitexin-treated obese mice. Additionally, vitexin inhibited NFκB expression by regulating IκBα phosphorylation, thereby alleviating obesity-induced liver injury. These findings suggest that vitexin may be the primary active component in hawthorn responsible for reducing blood lipid levels, highlighting its potential in the treatment of obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
- Bo Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ziqing Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lin Song
- Department of Pharmacy, Children' S Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Chen Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Changhong Shen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zefang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ruoqi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Rong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
17
|
Chen H, Qiu D, Miao X, Yang W, He Q, Ren H, Zhang L, Ruan H, Zhang J, Zhang N. Zearalenone Depresses Lactation Capacity Through the ROS-Mediated PI3K/AKT Pathway. Animals (Basel) 2025; 15:1050. [PMID: 40218442 PMCID: PMC11987832 DOI: 10.3390/ani15071050] [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: 02/12/2025] [Revised: 03/18/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
The effects of zearalenone (ZEA), a fungal toxin in food and feed, remain unclear on the mammary gland and lactation. This study examines ZEA-induced damage in lactating mice and bovine mammary epithelial cells (MAC-T), focusing on the role of the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway in regulating cell proliferation and apoptosis. The results demonstrated that exposure to ZEA at different doses (5 mg/kg, 10 mg/kg, and 20 mg/kg) reduced lactation in female mice and slowed weight gain in their offspring. Hematoxylin and eosin (HE) staining and CSNK immunofluorescence staining of mammary tissue confirmed ZEA-induced mammary gland damage in vivo. Further analysis using PCNA immunohistochemistry and fluorescent TUNEL staining revealed that ZEA promoted apoptosis and decreased the proliferative capacity of mammary tissues. In vitro, 20 μM ZEA decreased MAC-T cell proliferation, increased apoptosis and oxidative stress, inhibited PI3K/AKT signaling, and decreased κ-casein (CSNK) expression. Pretreatment with a reactive oxygen species (ROS) scavenger (NAC) or PI3K/AKT activator (740-Y-P) reversed these effects, with NAC specifically restoring PI3K/AKT activity inhibited by ZEA. Overall, this study concludes that ZEA induces MAC-T cell apoptosis and disrupts proliferation via the ROS-mediated PI3K/AKT pathway, ultimately impairing lactation function. These findings highlight potential targets for managing ZEA contamination in food and its impact on lactation.
Collapse
Affiliation(s)
- Hong Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.C.); (D.Q.); (X.M.); (W.Y.); (Q.H.); (H.R.); (L.Z.); (H.R.)
| | - Di Qiu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.C.); (D.Q.); (X.M.); (W.Y.); (Q.H.); (H.R.); (L.Z.); (H.R.)
| | - Xue Miao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.C.); (D.Q.); (X.M.); (W.Y.); (Q.H.); (H.R.); (L.Z.); (H.R.)
| | - Wenyue Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.C.); (D.Q.); (X.M.); (W.Y.); (Q.H.); (H.R.); (L.Z.); (H.R.)
| | - Qi He
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.C.); (D.Q.); (X.M.); (W.Y.); (Q.H.); (H.R.); (L.Z.); (H.R.)
| | - Hao Ren
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.C.); (D.Q.); (X.M.); (W.Y.); (Q.H.); (H.R.); (L.Z.); (H.R.)
| | - Luyao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.C.); (D.Q.); (X.M.); (W.Y.); (Q.H.); (H.R.); (L.Z.); (H.R.)
| | - Hongri Ruan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.C.); (D.Q.); (X.M.); (W.Y.); (Q.H.); (H.R.); (L.Z.); (H.R.)
| | - Jiantao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (H.C.); (D.Q.); (X.M.); (W.Y.); (Q.H.); (H.R.); (L.Z.); (H.R.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
| | - Na Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
18
|
Stokes AE, Clark HM, Edwards JL, Payton RR, Beever JE, Freeman TF, Hessock EA, Schrick FN, Moorey SE. Transcriptome profiles of blastocysts originating from oocytes matured in follicular fluid from preovulatory follicles of greater or lesser maturity. BMC Genomics 2025; 26:339. [PMID: 40186098 PMCID: PMC11969919 DOI: 10.1186/s12864-025-11521-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Oocyte competence for early embryo development relies on intercellular communication between the maturing oocyte and preovulatory follicle. Preovulatory follicle maturity, as indicated by serum estradiol concentration or follicle diameter, has previously been linked to pregnancy, follicular fluid metabolites, cumulus-oocyte metabolism, and oocyte competency for embryo development. Such relationships indicate metabolic and developmental programming of the oocyte based on the preovulatory follicle's physiological status, but downstream impacts on the molecular signature of blastocysts have not been examined. We hypothesized that supplementing maturing oocytes with follicular fluid originating from preovulatory follicles of greater or lesser maturity would impact the transcriptome of resulting blastocysts and indicate metabolic programming of the embryo that originated from the oocyte's maturation environment. The objective was to investigate the effect of follicle maturity on the oocyte by examining the transcriptome of blastocysts originating from oocytes matured in the presence of follicular fluid from preovulatory follicles of greater or lesser maturity. RESULTS In vitro maturing oocytes were supplemented with follicular fluid collected from preovulatory follicles of greater or lesser maturity. Following identical embryo culture procedures, RNA-sequencing was performed on pools of 2 blastocysts (Greater, n = 12; Lesser, n = 15; all with stage code = 7 and quality code = 1). A total of 12,310 genes were identified in blastocysts after filtering to remove lowly abundant genes. There were 113 genes that differed in expression between blastocysts originating from oocytes matured in greater versus lesser maturity follicular fluid (eFDR < 0.01). Although no pathways were significantly enriched with differentially expressed genes, transcriptome profiles suggested improved Wnt/β-catenin signaling, metabolism, and protection from oxidative stress in blastocysts derived from oocytes matured in greater maturity follicular fluid, while potential unregulated cell growth presented in blastocysts resulting from the lesser follicle maturity treatment. CONCLUSIONS Follicular fluid from preovulatory follicles of greater physiological maturity may better prepare maturing oocytes for early embryo development. Furthermore, oocytes matured in follicular fluid from preovulatory follicles of lesser maturity may attempt to overcompensate for nutrient deficit during oocyte maturation, leading to uncontrolled cellular growth and increased oxidative stress.
Collapse
Affiliation(s)
- Allyson E Stokes
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Hannah M Clark
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - J Lannett Edwards
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Rebecca R Payton
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Jon E Beever
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Trevor F Freeman
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Emma A Hessock
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - F Neal Schrick
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Sarah E Moorey
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA.
| |
Collapse
|
19
|
Zhou X, Chen W, Zhuang D, Xu G, Puyang Y, Rui H. Knockdown of SETD5 Inhibits Colorectal Cancer Cell Growth and Stemness by Regulating PI3K/AKT/mTOR Pathway. Biochem Genet 2025; 63:1924-1937. [PMID: 38641699 DOI: 10.1007/s10528-024-10766-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/28/2024] [Indexed: 04/21/2024]
Abstract
SET domain-containing 5 (SETD5), a member of protein lysine methyltransferase family, is expressed in multiple cancers, making it potential therapeutic targets. However, the role of SETD5 in colorectal cancer remains largely unknown. The expression of SETD5 in the 30 pairs colorectal cancer tissues samples and cell lines were determined by qRT-PCR. The functions of SETD5 was detected by knocked-down or overexpression in colorectal cancer cell lines SW480 and HCT116 cells. Cell proliferative activity, cell death, and stemness characteristics were assessed. BEZ235, a PI3K/AKT/mTOR pathway inhibitor, was used to perform rescue experiment to analyze whether SETD5 exerted its effects through activating PI3K/AKT/mTOR pathway. SETD5 was substantially upregulated in colorectal cancer, and correlated to metastasis and clinical stage of patients. Knockdown of SETD5 inhibited SW480 and HCT116 cell growth, as evidenced by the inhibition of cell viability and clone-forming. Moreover, Knockdown of SETD5 suppressed the capability of tumor sphere formation of SW480 and HCT116 cells, and reduced the expression of stemness-related proteins Nanog and Sox2. Further western blot analysis revealed that SETD5 knockdown inhibited the phosphorylation of proteins associated with the PI3K/AKT/mTOR pathway. In contrast, overexpression of SETD5 exerted the opposite effects. Mechanistically, by blocking PI3K/AKT/mTOR pathway with BEZ235, the effects of SETD5 overexpression on cell viability and Nanog and Sox2 protein expression were reversed. Our results substantiated that SETD5 functioned as an oncogene by promoting cell growth and stemness in colorectal cancer cells through activating the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaohua Zhou
- Department of General Surgery, Nanjing Gaochun People's Hospital, Gaochun, 211300, Jiangsu, China
| | - Wenqiang Chen
- Department of Medical Oncology, Nanjing Gaochun People's Hospital, Gaochun, 211300, Jiangsu, China
| | - Duanming Zhuang
- Department of Gastroenterology, Economic Development Zone, Nanjing Gaochun People's Hospital, No. 53, Maoshan, Gaochun, 211300, Jiangsu, China.
| | - Guangqi Xu
- Department of General Surgery, Nanjing Gaochun People's Hospital, Gaochun, 211300, Jiangsu, China
| | - Yongqiang Puyang
- Department of General Surgery, Nanjing Gaochun People's Hospital, Gaochun, 211300, Jiangsu, China
| | - Hongqing Rui
- Department of General Surgery, Nanjing Gaochun People's Hospital, Gaochun, 211300, Jiangsu, China
| |
Collapse
|
20
|
San Basilio M, Rodríguez-Laguna L, Triana Junco P, Martínez-Glez V, Ramirez-Amoros C, Delgado-Miguel C, Rodriguez-Arias JP, Lopez-Gutierrez JC. Association of Somatic KRAS Variants with Osteolysis in Arteriovenous Malformations. Eur J Pediatr Surg 2025. [PMID: 40157373 DOI: 10.1055/a-2540-3621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
The genetic study of vascular anomalies provides a better understanding of their etiopathogenesis and allows the use of targeted therapies. Activating KRAS pathogenic variants promote cell proliferation by activating MAPK and PI3K signalling pathways, which have been associated with the pathogenesis of vascular anomalies, especially high-flow ones such as arteriovenous malformations (AVMs). AVMs' genomic landscape is extensive, and a genotype-phenotype correlation has not been shown. In this study, we aimed to prove an association between KRAS gene mutations and the presence of osteolysis in patients with AVMs. Herein, we present a clinical-molecular retrospective study of patients with AVMs, bone involvement, and KRAS pathogenic variants.A retrospective review of patients with AVMs and KRAS somatic variants followed by the Vascular Anomalies Unit at our institution was performed. All patients present bone involvement. We analyzed demographics, clinical features (AVMs location, phenotype), treatment received, and response to treatment. Previous imaging studies were used to assess bone involvement. Genetic studies were performed by high-throughput sequencing using a custom-designed panel.Of the 77 patients with AVMs currently followed in our clinic, 60 (77.9%) had genetic testing, and 19 (31.6%) presented a KRAS somatic activating variant and were therefore included in the study. There were 12 women and 7 men aged 10 to 79 years. When studying radiographies or CT scans, we found that all 19 patients associated osteolysis adjacent to the AVMs. Regarding the KRAS variants, the most frequent one was p.Gly12Asp, followed by p.Gln61His and p.Gly13Arg. Additionally, we reviewed imaging studies from the other 41 patients with AVMs and different pathogenic variants such as MAP2K1, RASA1, and BRAF, and did not find osteolysis.We have described for the first time the relationship between somatic, activating KRAS pathogenic variants and osteolysis in patients with AVMs. Early detection of these KRAS alterations in this type of patient should lead us to rule out bone involvement. Moreover, identifying these mutations may help guide targeted therapies, potentially preventing the development of osteolysis and improving patient outcomes.
Collapse
Affiliation(s)
- María San Basilio
- Department of Pediatric Surgery, Hospital Universitario La Paz, Paseo de la Castellana, Madrid, Spain
| | | | - Paloma Triana Junco
- Department of Pediatric Surgery, Hospital Universitario La Paz, Paseo de la Castellana, Madrid, Spain
| | | | - Carla Ramirez-Amoros
- Department of Pediatric Surgery, Hospital Universitario La Paz, Paseo de la Castellana, Madrid, Spain
| | - Carlos Delgado-Miguel
- Department of Pediatric Surgery, Hospital Universitario La Paz, Paseo de la Castellana, Madrid, Spain
- Department of Pediatric Surgery, Hospital Universitario Fundacion Jimenez Diaz, Madrid, Spain
| | | | - Juan C Lopez-Gutierrez
- Division of Vascular Anomalies, Department of Pediatric Surgery, La Paz Children's Hospital, Madrid, Spain
| |
Collapse
|
21
|
Chen L, Luo D, Xiao H, Zeng Z, Luo H, Gao S, Tang X, Huang Z, Zeng Y. Mycoplasma genitalium protein of adhesion inhibits human urethral epithelial cells apoptosis via CypA/PI3K/AKT/mTOR-dependent autophagy. Front Microbiol 2025; 16:1570659. [PMID: 40207157 PMCID: PMC11979137 DOI: 10.3389/fmicb.2025.1570659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Background Mycoplasma genitalium, a prokaryotic microorganism, is a known pathogen of sexually transmitted infections. Previously, we identified cyclophilin A (CypA) as the membrane receptor on human urethral epithelial cells (SV-HUC-1) that binds to the M. genitalium protein of adhesion (MgPa) and demonstrated that recombinant MgPa (rMgPa) inhibits apoptosis via CypA-mediated regulation of the PI3K/AKT/NF-κB pathway. Given the established interplay between autophagy and apoptosis, this study aims to investigate whether rMgPa inhibits apoptosis in SV-HUC-1 cells by modulating CypA/PI3K/AKT/mTOR-dependent autophagy. Methods In this work, after SV-HUC-1 cells were stimulated with rMgPa, autophagy was detected using Western blotting, immunofluorescence and transmission electron microscopy, respectively. Western blotting and Annexin V/PI assays were used to determine the signaling pathway involved in rMgPa- inhibited apoptosis via inducing autophagy. Results rMgPa upregulated the autophagy-related proteins ATG7 and LC3B while downregulating P62 expression in SV-HUC-1 cells. Transmission electron microscopy showed the presence of intracellular autophagosomes, and indirect immunofluorescence confirmed the enhanced expression of LC3B, indicating that rMgPa induces autophagy. Silencing of CypA significantly attenuated rMgPa-induced autophagy, highlighting the essential role of CypA in this process. Furthermore, rMgPa was found to regulate the PI3K/AKT/mTOR pathway via CypA, thereby promoting autophagy. Western blot analysis and Annexin V/PI assays confirmed that rMgPa-induced autophagy inhibits apoptosis in urothelial cells through a CypA-dependent mechanism. Conclusion This study demonstrates that rMgPa suppresses apoptosis in SV-HUC-1 cells by inducing autophagy via CypA-mediated modulation of the PI3K/AKT/mTOR pathway, which elucidates a novel survival strategy employed by M. genitalium within host cells and provides valuable insights for potential therapeutic interventions targeting M. genitalium infections.
Collapse
Affiliation(s)
- Li Chen
- Basic Medical School, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, China
| | - Dan Luo
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Hua Xiao
- Basic Medical School, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, China
| | - Zhuo Zeng
- Basic Medical School, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, China
| | - Haodang Luo
- Basic Medical School, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, China
| | - Siqi Gao
- Basic Medical School, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, China
| | - Xiaoqian Tang
- Basic Medical School, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, China
| | - Zhijia Huang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yanhua Zeng
- Basic Medical School, Hengyang Medical College, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, China
| |
Collapse
|
22
|
Singh SR, Bhaskar R, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Mladenov M, Hadzi-Petrushev N, Stojchevski R, Sinha JK, Avtanski D. Exploring the Genetic Orchestra of Cancer: The Interplay Between Oncogenes and Tumor-Suppressor Genes. Cancers (Basel) 2025; 17:1082. [PMID: 40227591 PMCID: PMC11988167 DOI: 10.3390/cancers17071082] [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: 02/10/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Cancer is complex because of the critical imbalance in genetic regulation as characterized by both the overexpression of oncogenes (OGs), mainly through mutations, amplifications, and translocations, and the inactivation of tumor-suppressor genes (TSGs), which entail the preservation of genomic integrity by inducing apoptosis to counter the malignant growth. Reviewing the intricate molecular interplay between OGs and TSGs draws attention to their cell cycle, apoptosis, and cancer metabolism regulation. In the present review, we discuss seminal discoveries, such as Knudson's two-hit hypothesis, which framed the field's understanding of cancer genetics, leading to the next breakthroughs with next-generation sequencing and epigenetic profiling, revealing novel insights into OG and TSG dysregulation with opportunities for targeted therapy. The key pathways, such as MAPK/ERK, PI3K/AKT/mTOR, and Wnt/β-catenin, are presented in the context of tumor progression. Importantly, we further highlighted the advances in therapeutic strategies, including inhibitors of KRAS and MYC and restoration of TSG function, despite which mechanisms of resistance and tumor heterogeneity pose daunting challenges. A high-level understanding of interactions between OG-TSGs forms the basis for effective, personalized cancer treatment-something to strive for in better clinical outcomes. This synthesis should integrate foundational biology with translation and, in this case, contribute to the ongoing effort against cancer.
Collapse
Affiliation(s)
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan-si 38541, Republic of Korea;
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan-si 38541, Republic of Korea
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, India
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Symbiosis International (Deemed University), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, India
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | | | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
23
|
Alghamian Y, Soukkarieh C, Aljapawe A, Murad H. Exploring miRNA profile associated with cisplatin resistance in ovarian cancer cells. Biochem Biophys Rep 2025; 41:101906. [PMID: 39830525 PMCID: PMC11741906 DOI: 10.1016/j.bbrep.2024.101906] [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: 10/17/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Ovarian cancer is a common and lethal malignancy among women, whereas chemoresistance is one of the major challenges to its treatment and prognosis. Chemoresistance is a multifactorial phenomenon, involving various mechanisms that collectively modify the cell's response to treatment. Among the changes that arise in cells after acquiring chemoresistance is miRNA dysregulation. Here, this study aimed to identify miRNAs expression changes related to cisplatin resistance in ovarian cancer cells. The miRNA expression profiles of a cisplatin-sensitive A2780 cell line and two cisplatin-resistant cell lines, A2780cis and SK-OV-3, were analyzed using PCR array and qPCR. Accordingly, the miRNAs that were differentially expressed were further investigated to identify their biological functions and the target pathways using Gene Ontology (GO) annotation and KEGG pathway analyses. In order to evaluate the clinical significance of the differentially expressed miRNAs, survival analysis was carried out using expression data for ovarian cancer patients available in the Kaplan-Meier (KM) plotter database. The current work demonstrates that Nine miRNAs were found to be upregulated in cells resistant to cisplatin. Clearly, these miRNAs have functions in cell death/survival related processes and treatment response. They may also target pathways involved in treatment response like PI3K-Akt, pathway in cancer and MAPK. Interestingly, High expression of hsa-miR-133b, hsa-miR-512-are, hsa-miR-200b-3p, and hsa-miR-451a is related to poor overall survival in patients diagnosed with ovarian cancer. Our findings suggest that hsa-miR-133b, hsa-miR-512-5p, hsa-miR-200b-3p, and hsa-miR-451a are good candidates for future studies aimed to establishing functional links and exploring therapeutic interventions to overcome cisplatin resistance.
Collapse
Affiliation(s)
- Yaman Alghamian
- Department of Animal Biology, Faculty of Sciences, Damascus University, Syria
| | - Chadi Soukkarieh
- Department of Animal Biology, Faculty of Sciences, Damascus University, Syria
| | - Abdulmunim Aljapawe
- Department of Molecular and Biotechnology, Atomic Energy Commission of Syria (AECS), Syria
| | - Hossam Murad
- Department of Molecular and Biotechnology, Atomic Energy Commission of Syria (AECS), Syria
| |
Collapse
|
24
|
Zheng M, Yang X, Yuan P, Wang F, Guo X, Li L, Wang J, Miao S, Shi X, Ma S. Investigating the mechanism of Sinisan formula in depression treatment: a comprehensive analysis using GEO datasets, network pharmacology, and molecular docking. J Biomol Struct Dyn 2025; 43:2397-2411. [PMID: 38174416 DOI: 10.1080/07391102.2023.2297816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/14/2023] [Indexed: 01/05/2024]
Abstract
The herbal formula Sinisan (SNS) is a commonly used treatment for depression; however, its mechanism of action remains unclear. This article uses a combination of the GEO database, network pharmacology and molecular docking technologies to investigate the mechanism of action of SNS. The aim is to provide new insights and methods for future depression treatments. The study aims to extract effective compounds and targets for the treatment of depression from the T CMSP database. Relevant targets were searched using the GEO, Disgenet, Drugbank, PharmGKB and T T D databases, followed by screening of core targets. In addition, GO and KEGG pathway enrichment analyses were performed to explore potential pathways for the treatment of depression. Molecular docking was used to evaluate the potential targets and compounds and to identify the optimal core protein-compound complex. Molecular dynamics was used to further investigate the dynamic variability and stability of the complex. The study identified 118 active SNS components and 208 corresponding targets. Topological analysis of P P I networks identified 11 core targets. GO and KEGG pathway enrichment analyses revealed that the mechanism of action for depression involves genes associated with inflammation, apoptosis, oxidative stress, and the MAP K3 and P I3K-Akt signalling pathways. Molecular docking and dynamics simulations showed a strong binding affinity between these compounds and the screened targets, indicating promising biological activity. The present study investigated the active components, targets and pathways of SNS in the treatment of depression. Through a preliminary investigation, key signalling pathways and compounds were identified. These findings provide new directions and ideas for future research on the therapeutic mechanism of SNS and its clinical application in the treatment of depression.
Collapse
Affiliation(s)
- Meiling Zheng
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P.R. China
- Department of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P.R. China
| | - Xinxing Yang
- Department of Ultrasound, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P.R. China
| | - Ping Yuan
- Northwestern Polytechnical University Hospital, Xi'an, Shaanxi, P.R. China
| | - Feiyan Wang
- Department of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P.R. China
| | - Xiaodi Guo
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Long Li
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P.R. China
| | - Jin Wang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P.R. China
| | - Shan Miao
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P.R. China
| | - Xiaopeng Shi
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P.R. China
| | - Shanbo Ma
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
25
|
Huang L, Wang X, Zhou W, Li Z, Chen C, Sun Y. Hydrolyzed egg yolk peptide alleviates ovariectomy-induced osteoporosis by regulating lipid metabolism. Int J Biol Macromol 2025; 292:139223. [PMID: 39733873 DOI: 10.1016/j.ijbiomac.2024.139223] [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: 09/29/2024] [Revised: 11/28/2024] [Accepted: 12/24/2024] [Indexed: 12/31/2024]
Abstract
Osteoporosis is a systemic, progressive bone disease that causes metabolic disorders. Previous study identified the preventive effects of hydrolyzed egg yolk peptide (YPEP) on osteoporosis. However, the underlying antiosteoporosis mechanism remains unclear. Herein, 30 female rats were randomly divided into 5 groups (n = 6), including the sham, OVX, E2 (25 μg/kg/d 17β-estradiol), LYPEP (10 mg/kg/d YPEP), and HYPEP (40 mg/kg/d YPEP) groups. YPEP treatment significantly changed bone turnover marker levels and prevented the deterioration of bone structure and strength caused by ovariectomy. YPEP supplementation significantly changed endogenous metabolites related to lipid metabolism in the serum of ovariectomized rats, identifying 46 metabolites closely linked to bone biomarkers. Additionally, YPEP reduced the expression of the lipid metabolism-related protein peroxisome proliferator-activated receptor PPARγ and increased the expression of bone formation proteins BMP2 and RUNX2. Collectively, these results elucidated that YPEP improves osteoporosis by inhibiting lipogenesis to promote bone formation. This study provides novel evidence for the use of YPEP in treating osteoporosis.
Collapse
Affiliation(s)
- Ludi Huang
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xincen Wang
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Wei Zhou
- Radiology Department of Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), China
| | - Zeqi Li
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Chuanjing Chen
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yongye Sun
- School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
26
|
Zhang YJ, He H, Sawuer G, Ma XK, Ainiwaer Z, Wu DD, Zhang XX, An DQ. Tianxiangdan suppresses foam cell formation by enhancing lipophagy and reduces the progression of atherosclerosis. In Vitro Cell Dev Biol Anim 2025; 61:298-310. [PMID: 39808371 DOI: 10.1007/s11626-024-01004-z] [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: 06/13/2024] [Accepted: 11/10/2024] [Indexed: 01/16/2025]
Abstract
The aim of this study is to assess the impact of Tianxiangdan (TXD) on lipophagy in foam cells and its underlying mechanism in treating atherosclerosis, particularly focusing on its efficacy in lowering blood lipids. In vivo, ApoE-/- atherosclerosis mouse models were established for group intervention. Blood lipid levels of the mice were measured, lipid deposition and autophagy levels in atherosclerotic plaques were assessed, and co-localization of lipid droplets and autophagosomes was examined. In vitro, human THP-1 cells were induced into macrophages and then transformed into foam cells using ox-LDL induction. Different intervention groups were established. Total cellular cholesterol (TC), free cholesterol (FC), and autophagy levels were assessed, while the morphology and distribution of lipid droplets and autophagosomes in cells were observed using transmission electron microscopy. Western blot analysis was performed to evaluate the expression levels of PI3K, Akt, mTOR, TFEB, LC3II/I, ULK1, ABCA1, and p62. TXD effectively lowers blood lipid levels in ApoE-/- atherosclerotic mice, enhances lipophagy, and reduces lipid accumulation in foam cells and arterial lipid plaques. It achieves this by suppressing the expression of p85, Akt, and mTOR, while activating downstream autophagy signals such as TFEB, LC3II/I, and ULK1. Additionally, TXD reduces the expression of p62 and enhances the expression of the cholesterol transport molecule ABCA1. Our findings indicate that TXD activates lipophagy via the PI3K/Akt/mTOR pathway, leading to a reduction in lipid deposition within foam cells and plaques, thereby mitigating atherosclerosis.
Collapse
Affiliation(s)
- Ya-Jie Zhang
- College of Traditional Chinese Medicine, Xinjiang Uygur Autonomous Region, Xinjiang Medical University, Urumqi, 830063, China
| | - Huan He
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, China
| | - Guligena Sawuer
- Department of Cardiology, Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Xue-Kuan Ma
- Medical Department, Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Zulihumaer Ainiwaer
- College of Traditional Chinese Medicine, Xinjiang Uygur Autonomous Region, Xinjiang Medical University, Urumqi, 830063, China
| | - Dan-Dan Wu
- College of Traditional Chinese Medicine, Xinjiang Uygur Autonomous Region, Xinjiang Medical University, Urumqi, 830063, China
| | - Xia-Xia Zhang
- College of Traditional Chinese Medicine, Xinjiang Uygur Autonomous Region, Xinjiang Medical University, Urumqi, 830063, China
| | - Dong-Qing An
- College of Traditional Chinese Medicine, Xinjiang Uygur Autonomous Region, Xinjiang Medical University, Urumqi, 830063, China.
- Traditional Chinese Medicine Inheritance and Innovation Center, Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
- College of Traditional Chinese Medicine, Xinjiang Medical University, No. 567 of Shangde North Road, Shuimogou District, Urumqi, 830000, China.
| |
Collapse
|
27
|
Yang L, Hou H, Lu L, Sun Y, Chen R, Deng Q, Chen H. Effects of natural source polysaccharides on neurological diseases: A review. Int J Biol Macromol 2025; 296:139697. [PMID: 39805435 DOI: 10.1016/j.ijbiomac.2025.139697] [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: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
With the aging of society and changes in lifestyle, the incidence of neurological diseases (NDs) has been increasing year by year, bringing a heavy burden to patients and society. Although the efficacy of chemical drugs in the treatment of NDs is remarkable, there are problems such as high side effects and high costs. Therefore, finding mild and efficient drugs for NDs treatment has become an urgent clinical need. Natural source polysaccharides (NSPs) are macromolecules with unique bioactivity and low toxicity characteristics, which have great potential to become novel therapeutic agents for NDs. In the present study, the pharmacological activities and potential molecular mechanisms of NSPs to alleviate NDs are systematically reviewed from the perspectives of inflammation, oxidative stress, apoptosis, neuronal cell autophagy, neurotoxicity, and sedation-hypnosis. In addition, the limitations of the existing studies were analyzed and discussed, and the future research direction was suggested. This study may provide scientific basis for the research and development of therapeutic agents for NDs based on NSPs.
Collapse
Affiliation(s)
- Luyuan Yang
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Hailu Hou
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Liping Lu
- Guizhou Dalong Pharmaceutical Co., Ltd., Guiyang 550001, China
| | - Yu Sun
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Ruhai Chen
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Qingfang Deng
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Huaguo Chen
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China.
| |
Collapse
|
28
|
Silva-Pinto PA, de Pontes JTC, Aguilar-Morón B, Canales CSC, Pavan FR, Roque-Borda CA. Phytochemical insights into flavonoids in cancer: Mechanisms, therapeutic potential, and the case of quercetin. Heliyon 2025; 11:e42682. [PMID: 40084006 PMCID: PMC11904581 DOI: 10.1016/j.heliyon.2025.e42682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 03/16/2025] Open
Abstract
Quercetin, a flavonoid known for its potent antioxidant and anti-inflammatory properties, has gained attention in cancer therapy due to its ability to modulate key molecular pathways involved in tumor progression and immune evasion. This review provides a comprehensive analysis of quercetin's effects on pathways such as PI3K/Akt/mTOR, MAPK/ERK, NF-κB, and JAK/STAT, which are central to cancer cell survival, proliferation, and apoptosis. Through inhibition of PI3K/Akt/mTOR and MAPK/ERK signaling, quercetin promotes apoptosis and reduces proliferation specifically in cancer cells while sparing healthy cells. Additionally, quercetin downregulates NF-κB activity and modulates JAK/STAT signaling, enhancing immune recognition of cancer cells and decreasing inflammation in the tumor microenvironment. Emerging nanoformulation strategies are also discussed, highlighting how nanotechnology can improve quercetin's bioavailability and targeting capabilities. Unlike other reviews, this work uniquely integrates molecular insights with cutting-edge nanoformulations, showcasing quercetin's dual potential as a therapeutic agent and an immune modulator in the evolving landscape of cancer treatment. This review underscores quercetin's multifaceted role in cancer treatment and suggests future directions to optimize its clinical efficacy, particularly in combination with conventional therapies.
Collapse
Affiliation(s)
- Piero Alex Silva-Pinto
- Vicerrectorado de Investigación, Universidad Católica de Santa María de Arequipa, Arequipa, 04000, Republic of Peru
| | - Janaína Teixeira Costa de Pontes
- Department of Biological Sciences, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-900, SP, Brazil
| | - Brigitte Aguilar-Morón
- Facultad de Ingeniería de Procesos – Universidad Nacional de San Agustín, Arequipa, Arequipa, Republic of Peru
| | | | - Fernando Rogério Pavan
- Department of Biological Sciences, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, 14800-900, SP, Brazil
| | - Cesar Augusto Roque-Borda
- Vicerrectorado de Investigación, Universidad Católica de Santa María de Arequipa, Arequipa, 04000, Republic of Peru
| |
Collapse
|
29
|
Liu J, Qiu L, Chen J, Zeng T. Lycorine hydrochloride Suppresses the Proliferation and Invasion of Esophageal Cancer by Targeting TRIM22 and Inhibiting the JAK2/STAT3 and Erk Pathways. Cancers (Basel) 2025; 17:718. [PMID: 40075566 PMCID: PMC11898953 DOI: 10.3390/cancers17050718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Tumor metastasis and poor drug efficacy are two of the most common causes of therapeutic failure in cancer patients. The underlying molecular mechanism requires further exploration, and novel effective curative strategies are urgently needed. Nature is a rich source of novel drugs, and Lycorine hydrochloride (Lyc.HCL) is a natural alkaloid with tremendous therapeutic potential. However, the molecular mechanisms of its antitumor activity are still unknown. In the current study, we investigated the effects and mechanisms of Lyc.HCL against esophageal squamous cell carcinomas (ESCCs), which pose serious threats to human life. METHODS An MTS assay and a clone formation assay were used to assess the viability of ESCC cell lines after Lyc.HCL treatment in vitro. Apoptosis and cell cycle regulation were analyzed using flow cytometry. Wound healing and Transwell assays were used to analyze cell migration, while invasion was analyzed using the Matrigel Transwell assay. We detected the expression of tripartite motif-containing 22 (TRIM22) through immunohistochemistry and Western blotting. A docking experiment was performed to explore the targets of Lyc.HCL. The expression levels of Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/extracellular signal-regulated kinase (Erk) pathway components were detected through Western blotting. A rescue experiment was performed to determine the potential role of TRIM22. In addition, we explored the in vivo anti-ESCC effects and mechanism of Lyc.HCL by using it to treat tumor-bearing mice. RESULTS The Lyc.HCL treatment was found to inhibit esophageal squamous cell carcinoma cell proliferation both in vitro and in vivo by blocking the cell cycle at the G2 phase, inhibiting cell migration and invasion. We found that the TRIM22 protein was highly expressed in ESCCs but not in normal esophageal tissue. Lyc.HCL directly targeted TRIM22, decreasing the expression of TRIM22 and the JAK2/STAT3 and Erk signaling pathways, both in vitro and in vivo. Using animal experiments, we observed that the depletion of TRIM22 delayed tumor growth, but this effect was significantly reversed upon TRIM22 overexpression. CONCLUSIONS Taken together, these findings demonstrate that Lyc.HCL can effectively suppress ESCC both in vitro and in vivo by targeting TRIM22 and regulating the JAK2/STAT3 and Erk pathways. These results suggest that Lyc.HCL may serve as a potential novel therapeutic for ESCC, with TRIM22 emerging as a promising target for treatment.
Collapse
Affiliation(s)
- Jingyan Liu
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524023, China
- Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Liangxian Qiu
- Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jialing Chen
- Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Tao Zeng
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524023, China
| |
Collapse
|
30
|
Qiang M, Chen Z, Liu H, Dong J, Gong K, Zhang X, Huo P, Zhu J, Shao Y, Ma J, Zhang B, Liu W, Tang M. Targeting the PI3K/AKT/mTOR pathway in lung cancer: mechanisms and therapeutic targeting. Front Pharmacol 2025; 16:1516583. [PMID: 40041495 PMCID: PMC11877449 DOI: 10.3389/fphar.2025.1516583] [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: 10/24/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
Owing to its high mortality rate, lung cancer (LC) remains the most common cancer worldwide, with the highest malignancy diagnosis rate. The phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling (PAM) pathway is a critical intracellular pathway involved in various cellular functions and regulates numerous cellular processes, including growth, survival, proliferation, metabolism, apoptosis, invasion, and angiogenesis. This review aims to highlight preclinical and clinical studies focusing on the PAM signaling pathway in LC and underscore the potential of natural products targeting it. Additionally, this review synthesizes the existing literature and discusses combination therapy and future directions for LC treatment while acknowledging the ongoing challenges in the field. Continuous development of novel therapeutic agents, technologies, and precision medicine offers an increasingly optimistic outlook for the treatment of LC.
Collapse
Affiliation(s)
- Min Qiang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Zhe Chen
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hongyang Liu
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Junxue Dong
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Kejian Gong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xinjun Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Peng Huo
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jingjun Zhu
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yifeng Shao
- Department of General Surgery, Capital Institute of Pediatrics’ Children’s Hospital, Beijing, China
| | - Jinazun Ma
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Bowei Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Mingbo Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
31
|
Sicurella M, De Chiara M, Neri LM. Hedgehog and PI3K/Akt/mTOR Signaling Pathways Involvement in Leukemic Malignancies: Crosstalk and Role in Cell Death. Cells 2025; 14:269. [PMID: 39996741 PMCID: PMC11853774 DOI: 10.3390/cells14040269] [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: 01/21/2025] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/26/2025] Open
Abstract
The Hedgehog (Hh) and PI3K/Akt/mTOR signaling pathways play a pivotal role in driving the initiation and progression of various cancers, including hematologic malignancies such as acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), and chronic lymphocytic leukemia (CLL). These pathways are often dysregulated in leukemia cells, leading to increased cell growth, survival, and drug resistance while also impairing mechanisms of cell death. In leukemia, the Hh pathway can be abnormally activated by genetic mutations. Additionally, the PI3K/Akt/mTOR pathway is frequently overactive due to genetic changes. A key aspect of these pathways is their interaction: activation of the PI3K/Akt pathway can trigger a non-canonical activation of the Hh pathway, which further promotes leukemia cell growth and survival. Targeted inhibitors of these pathways, such as Gli inhibitors and PI3K/mTOR inhibitors, have shown promise in preclinical and clinical studies.
Collapse
Affiliation(s)
- Mariaconcetta Sicurella
- Department of Environmental Sciences and Prevention, University of Ferrara, 44121 Ferrara, Italy;
| | - Marica De Chiara
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy;
| | - Luca Maria Neri
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy;
- LTTA-Electron Microscopy Center, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
32
|
Xin L, Feng HC, Zhang Q, Cen XL, Huang RR, Tan GY, Zhang Q. Exploring the osteogenic effects of simiao wan through activation of the PI3K/AKT pathway in osteoblasts. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119023. [PMID: 39489361 DOI: 10.1016/j.jep.2024.119023] [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: 08/09/2024] [Revised: 10/21/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Osteoporosis (OP) is a degenerative bone disease commonly associated with reduced bone density and increased fracture risk. AIM OF THE STUDY This study aimed to validate the therapeutic effects of Simiao wan (SMW) on OP and explore the underlying mechanism, particularly focusing on the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway. MATERIALS AND METHODS The chemical components of SMW were identified using UPLC-Q-TOF-MS/MS. The obtained compounds were then input into the TCMSP, TargetNet, and SwissTargetPrediction databases to predict potential targets. OP-related targets were collected from the GeneCards and DisGeNET databases, and intersecting targets were identified through a Venn diagram. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed on the intersecting targets using the Database for Annotation, Visualization and Integrated Discovery (DAVID). SMW extract was subsequently used to treat osteoblasts in vitro, and its toxicity on osteoblasts was assessed using Cell Counting Kit-8 (CCK-8) and lactate dehydrogenase (LDH) release assays. Osteoblast differentiation and activity were further evaluated using alizarin red staining, alkaline phosphatase staining, and Western blot analyses to validate the activation of network pharmacological signaling pathways. RESULTS A total of 121 potential targets were identified for SMW in the treatment of OP, with AKT1 as the primary target. The PI3K/AKT pathway emerged as a key signaling pathway potentially involved in SMW's therapeutic effects o OP. Toxicity assessments showed no significant toxicity of SMW on osteoblasts. Additionally, SMW promoted osteoblast proliferation, alkaline phosphatase activity, calcium nodule deposition, and the expression of osteogenic markers (osteocalcin (OCN), runt-related transcription factor 2 (RunX2), and collagen I), and activated the PI3K/AKT signaling pathway. The PI3K/AKT pathway inhibitor LY294002 partially reversed the SMW-induced mineral deposition and expression of OCN, RunX2, and collagen I. CONCLUSION SMW demonstrated effective multi-target and multi-pathway therapeutic potential in the treatment of OP, with a significant impact on the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Li Xin
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China; Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Huan-Cun Feng
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Qiang Zhang
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Xiao-Lin Cen
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Rong-Rong Huang
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Guo-Yao Tan
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Qun Zhang
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
33
|
Wu Z, Yu L, Hu Y, Bao W, Wu S. Paeoniflorin Inhibits Porcine Circovirus Type 2 Replication by Inhibiting Autophagy and Targeting AKT/mTOR Signaling. Vet Sci 2025; 12:117. [PMID: 40005877 PMCID: PMC11860941 DOI: 10.3390/vetsci12020117] [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: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
Porcine circovirus type 2 (PCV2) is an important pathogen that leads to great economic losses to the swine industry. Paeoniflorin (PF), a novel plant extract, has been reported to have antiviral properties. However, the role of paeoniflorin in regulating PCV2 replication remains unclear. Here, we used the CCK8 assay to demonstrate that PF within safe concentrations (0-275 mM) significantly inhibits PCV2 replication in a dose-dependent manner in porcine kidney cells. Subsequently, comparative transcriptome and functional verification revealed that PF probably inherits PCV2 replication via targeting AKT/mTOR signaling. Further experimental data show that the AKT/mTOR signaling pathway is highly relevant to autophagy. Thus, experimental data from Western blot, qPCR, and the indirect immunofluorescence test indicate that PF inhibits PCV2 replication by inhibiting autophagy by targeting the AKT/mTOR signaling pathway. Together, our results provide insight into the mechanism of paeoniflorin in regulating PCV2 replication and offer new ideas for the treatment of PCV2 infection in pigs.
Collapse
Affiliation(s)
- Zhengchang Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.W.); (L.Y.); (Y.H.); (W.B.)
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Luchen Yu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.W.); (L.Y.); (Y.H.); (W.B.)
| | - Yueqing Hu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.W.); (L.Y.); (Y.H.); (W.B.)
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.W.); (L.Y.); (Y.H.); (W.B.)
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.W.); (L.Y.); (Y.H.); (W.B.)
| |
Collapse
|
34
|
Liu Y, Liu M, He X, Yang L, Zhang M, Tang P, Xing L, Niu H, Wang H. Molecular landscape of CD8 + T cells in pure red cell aplasia. Ann Hematol 2025; 104:953-961. [PMID: 39888354 PMCID: PMC11971129 DOI: 10.1007/s00277-025-06220-5] [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: 02/01/2025]
Abstract
The aberrant function of lymphocytes is considered a significant contributing factor to pure red cell aplasia (PRCA), but the precise mechanism by which T lymphocytes induce erythroid development stagnation remains unclear. In our study, the CD8+ T lymphocytes were isolated from bone marrow aspirates of acquired PRCA patients and healthy controls. RNA sequencing (RNA-Seq) was performed to analyze gene expression profiles. Additionally, the expression levels of key molecules and transcription factors were assessed at the transcription and protein levels. The RNA-Seq analysis revealed a significant upregulation of genes associated with the PI3K/AKT/mTOR pathway in CD8+ T lymphocytes from patients with PRCA, compared to healthy controls. The mRNA expression of AKT, mTOR and key transcription factors T-bet were significantly upregulated in CD8+ T cells from patients with PRCA. Treatment with rapamycin, an mTOR inhibitor, attenuated the activation of CD8+ T lymphocytes in PRCA patients. Our findings demonstrate the activation of the PI3K/AKT/mTOR signaling pathway in CD8+ T lymphocytes of PRCA patients, suggesting its involvement in PRCA pathogenesis. Targeting this pathway may offer a potential therapeutic strategy for PRCA characterized by CD8+ T cell dysregulation.
Collapse
Affiliation(s)
- Yumei Liu
- Department of Hematology, Tianjin Medical University General Hospital, No. 154 Anshandao Road, Heping District, Tianjin, 300052, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, 300052, China
- Tianjin Institute of Hematology, Tianjin, 300052, China
| | - Mengyuan Liu
- Department of Hematology, Tianjin Medical University General Hospital, No. 154 Anshandao Road, Heping District, Tianjin, 300052, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, 300052, China
- Tianjin Institute of Hematology, Tianjin, 300052, China
| | - Xiaoman He
- Department of Hematology, Tianjin Medical University General Hospital, No. 154 Anshandao Road, Heping District, Tianjin, 300052, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, 300052, China
- Tianjin Institute of Hematology, Tianjin, 300052, China
| | - Liyan Yang
- Department of Hematology, Tianjin Medical University General Hospital, No. 154 Anshandao Road, Heping District, Tianjin, 300052, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, 300052, China
- Tianjin Institute of Hematology, Tianjin, 300052, China
| | - Mengying Zhang
- Department of Hematology, Tianjin Medical University General Hospital, No. 154 Anshandao Road, Heping District, Tianjin, 300052, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, 300052, China
- Tianjin Institute of Hematology, Tianjin, 300052, China
| | - Pu Tang
- Department of Hematology, Tianjin Medical University General Hospital, No. 154 Anshandao Road, Heping District, Tianjin, 300052, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, 300052, China
- Tianjin Institute of Hematology, Tianjin, 300052, China
| | - Limin Xing
- Department of Hematology, Tianjin Medical University General Hospital, No. 154 Anshandao Road, Heping District, Tianjin, 300052, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, 300052, China
- Tianjin Institute of Hematology, Tianjin, 300052, China
| | - Haiyue Niu
- Department of Hematology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| | - Huaquan Wang
- Department of Hematology, Tianjin Medical University General Hospital, No. 154 Anshandao Road, Heping District, Tianjin, 300052, China.
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, 300052, China.
- Tianjin Institute of Hematology, Tianjin, 300052, China.
| |
Collapse
|
35
|
Maqbool MF, Gul S, Ishaq M, Maryam A, Khan M, Shakir HA, Irfan M, Li Y, Ma T. Theabrownin: a dietary nutraceutical with diverse anticancer mechanisms. Nat Prod Res 2025; 39:817-833. [PMID: 38284642 DOI: 10.1080/14786419.2024.2306917] [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: 09/14/2023] [Revised: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 01/30/2024]
Abstract
Cancer, a highly deadly disease, necessitates safe, cost-effective, and readily accessible treatments to mitigate its impact. Theabrownin (THBR), a polyphenolic pigment found in Pu-erh tea, has garnered attention for its potential benefits in memory, liver health, and inflammation control. By observing different biological activities of THBR, recently researchers have unveiled THBR's promising anticancer properties across various human cancer types. By examining existing studies, it is evident that THBR demonstrates substantial potential in inhibiting cell proliferation and reducing tumour size with minimal harm to normal cells. These effects are achieved through the modulation of key molecular markers such as Bcl-2, Bax, various Caspases, Poly (ADP-ribose) polymerase cleavage (Cl-PARP), and zinc finger E box binding homeobox 1 (ZEB 1). This review aims to provide in-depth insights into THBR's role in cancer research. This review also elucidates the underlying anticancer mechanisms of THBR, offering promise as a novel anticancer drug to alleviate the global cancer burden.
Collapse
Affiliation(s)
- Muhammad Faisal Maqbool
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Sameena Gul
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Ishaq
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Amara Maryam
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Khan
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Hafiz Abdullah Shakir
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Yongming Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tonghui Ma
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
36
|
An Y, Cao S, Shi L, Zhang Y, Wang X, Yuan S, Shi Y, Wang B, Liu J, Han CJ. Pharmacological modulation of Sigma-1 receptor ameliorates pathological neuroinflammation in rats with diabetic neuropathic pain via the AKT/GSK-3β/NF-κB pathway. Brain Res Bull 2025; 221:111226. [PMID: 39870326 DOI: 10.1016/j.brainresbull.2025.111226] [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: 11/03/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 01/29/2025]
Abstract
Diabetic neuropathic pain (DNP) is a common complication of diabetes mellitus (DM) and is characterized by spontaneous pain and neuroinflammation. The Sigma-1 receptor (Sig-1R) has been proposed as a target for analgesic development. It is an important receptor with anti-inflammatory properties and has been found to regulate DNP. However, it is not known whether Sig-1R can ameliorate pathological neuroinflammation in DNP. The present study used a rat model of DNP and a highly selective agonist of Sig-1R to assess the effects of the protein on neuropathic pain in rats with type 2 diabetes mellitus. The rats were divided into Control, Model, Sig-1R agonist PRE-084 (0.3, 0.6, 1 mg/kg), and metformin (Met, 20 mg/kg) groups, with seven rats per group, and their body weight, fasting blood glucose, mechanical withdrawal threshold and thermal withdrawal latency were tested weekly for two weeks. After treatment with PRE-084, the pain thresholds in the DNP rats were significantly improved, together with pathological changes in the dorsal root ganglion, reductions in the serum levels of TNF-α, IL-1β, IL-6, MOD, and prostaglandin E2 (PGE2), and the activity of superoxide dismutase was increased. The mRNA levels of TNF-α, IL-1β, and cyclooxygenase 2 (COX-2) were reduced. Pharmacological inhibition of Sig-1R with BD1047 (10 μM) abolished Sig-1R-mediated activation of lipopolysaccharide-treated BV-2 microglial cells. It was also found that PRE-084 increased phosphorylation of serine/threonine protein kinase B (AKT) and glycogen synthase kinase 3β (GSK-3β) at Ser9, inhibiting nuclear factor kappa B (NF-κB)-mediated neuroinflammation in the dorsal root ganglion, thus reducing DNP. The findings suggest that the effect of Sig-1R agonist PRE-084 on DNP may reduce the level of inflammation through the up-regulation of AKT/GSK-3β and down-regulation of the NF-κB signaling, thereby contributing to the treatment of the disease.
Collapse
Affiliation(s)
- Yuyu An
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Shanshan Cao
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Leilei Shi
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Yuhan Zhang
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Xin Wang
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Shiyu Yuan
- Department of Pharmacy, The Second affiliated hospital of Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Yongheng Shi
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xianyang 712046, PR China.
| | - Bin Wang
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xianyang 712046, PR China.
| | - Jiping Liu
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xianyang 712046, PR China.
| | - Chao-Jun Han
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xianyang 712046, PR China.
| |
Collapse
|
37
|
Megid RA, Ribeiro GG, Gomes INF, Laus AC, Ferro Leal L, Sussuchi da Silva L, Ariwoola ABA, Dias JM, Reis RM, Jose da Silva-Oliveira R. Sotorasib resistance triggers epithelial-mesenchymal transition and activates AKT and P38-mediated signaling. Front Mol Biosci 2025; 12:1537523. [PMID: 39950162 PMCID: PMC11821485 DOI: 10.3389/fmolb.2025.1537523] [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: 12/01/2024] [Accepted: 01/14/2025] [Indexed: 02/16/2025] Open
Abstract
Background The molecular non-genetic changes of resistance to sotorasib are currently uncertain. The aim of this study was to generate a sotorasib-resistant cell line via selective pressure and systematically examine the molecular and phenotypic alterations caused by resistance. Methods Mutant NCI-H358 (KRASG12C) were exposed to incremental doses (2-512 nM) of sotorasib. Then, resistant clones were separated by single-cell sorting. Proliferation was analyzed in real-time by xCELLigence; protein profiles were quantified by protein arrays; and mRNA expression profile was measured using the PanCancer Pathways panel by NanoString. In silico analyses were conducted from a database comprising patient-derived xenograft (PDX) models and cell lines resistant to sotorasib. AKT and p38. The synergistic effect of combining AKT, p38, and EGFR inhibitors was assessed using the SynergyFinder platform. Additionally, AKT and p38 genes were silenced using esiRNA. Results Sotorasib-resistant H358-R cell line displayed markers of the mesenchymal-epithelial transition and loss of cell adhesion. Were identified 30 overexpressed genes in the resistance model, implicating in signaling pathways that leads to AKT activation and heightened protein expression levels of phosphorylated AKT and p38. To identify potential therapeutic strategies for overcoming sotorasib resistance, we investigated the combination of AKT and p38 inhibitors. Notably, combined inhibition of AKT (MK2206) and p38 (adezmapimod) restored sensitivity to sotorasib in resistant cell lines, as did silencing AKT expression. Conclusion These findings underscore the importance of adaptive mechanisms in sotorasib resistance in NSCLC cells contributing by EMT activation and demonstrates synergic combination with AKT and p38 inhibitors to restore sotorasib sensitivity in KRASG12C cells.
Collapse
Affiliation(s)
| | | | | | - Ana Carolina Laus
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
| | - Letícia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
- Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, São Paulo, Brazil
| | | | | | | | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
- Life and Health Sciences Research Institute (ICVS) Medical School, University of Minho, Braga, Portugal
| | - Renato Jose da Silva-Oliveira
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
- Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, São Paulo, Brazil
| |
Collapse
|
38
|
Korytina GF, Markelov VA, Gibadullin IA, Zulkarneev SR, Nasibullin TR, Zulkarneev RH, Avzaletdinov AM, Avdeev SN, Zagidullin NS. The Relationship Between Differential Expression of Non-coding RNAs (TP53TG1, LINC00342, MALAT1, DNM3OS, miR-126-3p, miR-200a-3p, miR-18a-5p) and Protein-Coding Genes (PTEN, FOXO3) and Risk of Idiopathic Pulmonary Fibrosis. Biochem Genet 2025:10.1007/s10528-024-11012-z. [PMID: 39881079 DOI: 10.1007/s10528-024-11012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a rapidly progressive interstitial lung disease of unknown pathogenesis with no effective treatment currently available. Given the regulatory roles of lncRNAs (TP53TG1, LINC00342, H19, MALAT1, DNM3OS, MEG3), miRNAs (miR-218-5p, miR-126-3p, miR-200a-3p, miR-18a-5p, miR-29a-3p), and their target protein-coding genes (PTEN, TGFB2, FOXO3, KEAP1) in the TGF-β/SMAD3, Wnt/β-catenin, focal adhesion, and PI3K/AKT signaling pathways, we investigated the expression levels of selected genes in peripheral blood mononuclear cells (PBMCs) and lung tissue from patients with IPF. Lung tissue and blood samples were collected from 33 newly diagnosed, treatment-naive patients and 70 healthy controls. Gene expression levels were analyzed by RT-qPCR. TaqMan assays and TaqMan MicroRNA assay were employed to quantify the expression of target lncRNAs, mRNAs, and miRNAs. Our study identified significant differential expression in PBMCs from IPF patients compared to healthy controls, including lncRNAs MALAT1 (Fold Change = 3.809, P = 0.0001), TP53TG1 (Fold Change = 0.4261, P = 0.0021), and LINC00342 (Fold Change = 1.837, P = 0.0448); miRNAs miR-126-3p (Fold Change = 0.102, P = 0.0028), miR-200a-3p (Fold Change = 0.442, P = 0.0055), and miR-18a-5p (Fold Change = 0.154, P = 0.0034); and mRNAs FOXO3 (Fold Change = 4.604, P = 0.0032) and PTEN (Fold Change = 2.22, P = 0.0011). In lung tissue from IPF patients, significant expression changes were observed in TP53TG1 (Fold Change = 0.2091, P = 0.0305) and DNM3OS (Fold Change = 4.759, P = 0.05). Combined analysis of PBMCs expression levels for TP53TG1, MALAT1, miRNA miR-126-3p, and PTEN distinguished IPF patients from healthy controls with an AUC = 0.971, sensitivity = 0.80, and specificity = 0.955 (P = 6 × 10-8). These findings suggest a potential involvement of the identified ncRNAs and mRNAs in IPF pathogenesis. However, additional functional validation studies are needed to elucidate the precise molecular mechanisms by which these lncRNAs, miRNAs, and their targets contribute to PF.
Collapse
Affiliation(s)
- Gulnaz F Korytina
- Institute of Biochemistry and Genetics-Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences (IBG UFRC RAS), Pr. Oktyabrya, 71, Ufa, 450054, Russian Federation.
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation.
| | - Vitaly A Markelov
- Institute of Biochemistry and Genetics-Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences (IBG UFRC RAS), Pr. Oktyabrya, 71, Ufa, 450054, Russian Federation
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | - Irshat A Gibadullin
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | - Shamil R Zulkarneev
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | - Timur R Nasibullin
- Institute of Biochemistry and Genetics-Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences (IBG UFRC RAS), Pr. Oktyabrya, 71, Ufa, 450054, Russian Federation
| | - Rustem H Zulkarneev
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | | | - Sergey N Avdeev
- Sechenov First Moscow State Medical University (Sechenov University), 8-2, Trubetskaya Str., Moscow, 119991, Russian Federation
| | - Naufal Sh Zagidullin
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| |
Collapse
|
39
|
Xing J, Feng X, Zhang R, Zhang K. Targeting Hepatocellular Carcinoma Growth: Haprolid's Inhibition of AKT Signaling Through DExH-Box Helicase 9 Downregulation. Cancers (Basel) 2025; 17:443. [PMID: 39941810 PMCID: PMC11816161 DOI: 10.3390/cancers17030443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/19/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Objective: Haprolid, a novel compound extracted from Myxobacterium, has been proven to possess selective toxicity towards various tumor cells, effectively inhibiting the growth of hepatocellular carcinoma (HCC). However, the underlying molecular mechanism remains unclear. Methods: To identify differentially expressed proteins (DEPs), isobaric tags for relative and absolute quantitation (iTRAQ) were employed. The clinical significance of DExH-Box Helicase 9 (DHX9) was determined using tissue microarrays in HCC patients. Changes in protein expression were detected using Western blotting, qPCR, and immunohistochemistry. Cell proliferation was evaluated using CCK-8 and crystal violet staining. Cell apoptosis was assessed using Alexa Fluor 647 Annexin V. Xenograft tumor experiments were conducted in animals. Results: iTRAQ screening identified DHX9 as a DEP. DHX9 was discovered to be highly expressed in HCC tissues, correlating with poor prognosis in patients. Haprolid downregulated DHX9 expression, while knockdown of DHX9 suppressed HCC cell proliferation and migration and promoted apoptosis. Meanwhile, overexpression of DHX9 mitigated the inhibitory effect of Haprolid on HCC cells. Knockdown of DHX9 inhibited the AKT signaling pathway, and SC79 reversed the inhibitory effect of DHX9 knockdown on HCC cells. Xenograft experiments confirmed that the knockdown of DHX9 inhibited HCC growth, while the overexpression of DHX9 attenuated the inhibitory effect of Haprolid on HCC growth. Conclusions: Haprolid inhibits the AKT signaling pathway by downregulating DHX9, ultimately suppressing HCC growth. This finding opens up new avenues for targeted HCC therapy.
Collapse
Affiliation(s)
| | | | | | - Kaiguang Zhang
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (J.X.)
| |
Collapse
|
40
|
Kes MMG, Morales-Rodriguez F, Zaal EA, de Souza T, Proost N, van de Ven M, van den Heuvel-Eibrink MM, Jansen JWA, Berkers CR, Drost J. Metabolic profiling of patient-derived organoids reveals nucleotide synthesis as a metabolic vulnerability in malignant rhabdoid tumors. Cell Rep Med 2025; 6:101878. [PMID: 39708810 PMCID: PMC11866552 DOI: 10.1016/j.xcrm.2024.101878] [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: 11/22/2023] [Revised: 08/27/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024]
Abstract
Malignant rhabdoid tumor (MRT) is one of the most aggressive childhood cancers for which no effective treatment options are available. Reprogramming of cellular metabolism is an important hallmark of cancer, with various metabolism-based drugs being approved as a cancer treatment. In this study, we use patient-derived tumor organoids (tumoroids) to map the metabolic landscape of several pediatric cancers. Combining gene expression analyses and metabolite profiling using mass spectrometry, we find nucleotide biosynthesis to be a particular vulnerability of MRT. Treatment of MRT tumoroids with de novo nucleotide synthesis inhibitors methotrexate (MTX) and BAY-2402234 lowers nucleotide levels in MRT tumoroids and induces apoptosis. Lastly, we demonstrate in vivo efficacy of MTX in MRT patient-derived xenograft (PDX) mouse models. Our study reveals nucleotide biosynthesis as an MRT-specific metabolic vulnerability, which can ultimately lead to better treatment options for children suffering from this lethal pediatric malignancy.
Collapse
Affiliation(s)
- Marjolein M G Kes
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Francisco Morales-Rodriguez
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Esther A Zaal
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Terezinha de Souza
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Natalie Proost
- Preclinical Intervention Unit of the Mouse Clinic for Cancer and Ageing (MCCA), Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marieke van de Ven
- Preclinical Intervention Unit of the Mouse Clinic for Cancer and Ageing (MCCA), Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Division of Child Health, Wilhelmina Children's Hospital, Utrecht University, Utrecht, the Netherlands
| | - Jeroen W A Jansen
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Celia R Berkers
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| |
Collapse
|
41
|
Oladapo A, Deshetty UM, Callen S, Buch S, Periyasamy P. Single-Cell RNA-Seq Uncovers Robust Glial Cell Transcriptional Changes in Methamphetamine-Administered Mice. Int J Mol Sci 2025; 26:649. [PMID: 39859365 PMCID: PMC11766323 DOI: 10.3390/ijms26020649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/07/2025] [Accepted: 01/12/2025] [Indexed: 01/30/2025] Open
Abstract
Methamphetamine is a highly addictive stimulant known to cause neurotoxicity, cognitive deficits, and immune dysregulation in the brain. Despite significant research, the molecular mechanisms driving methamphetamine-induced neurotoxicity and glial cell dysfunction remain poorly understood. This study investigates how methamphetamine disrupts glial cell function and contributes to neurodevelopmental and neurodegenerative processes. Using single-cell RNA sequencing (scRNA-seq), we analyzed the transcriptomes of 4000 glial cell-associated genes from the cortical regions of mice chronically administered methamphetamine. Methamphetamine exposure altered the key pathways in astrocytes, including the circadian rhythm and cAMP signaling; in microglia, affecting autophagy, ubiquitin-mediated proteolysis, and mitophagy; and in oligodendrocytes, disrupting lysosomal function, cytoskeletal regulation, and protein processing. Notably, several transcription factors, such as Zbtb16, Hif3a, Foxo1, and Klf9, were significantly dysregulated in the glial cells. These findings reveal profound methamphetamine-induced changes in the glial transcriptomes, particularly in the cortical regions, highlighting potential molecular pathways and transcription factors as targets for therapeutic intervention. This study provides novel insights into the glial-mediated mechanisms of methamphetamine toxicity, contributing to our understanding of its effects on the central nervous system and laying the groundwork for future strategies to mitigate its neurotoxic consequences.
Collapse
Affiliation(s)
| | | | | | | | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.O.); (U.M.D.); (S.C.); (S.B.)
| |
Collapse
|
42
|
Lu C, Wei J, Gao C, Sun M, Dong D, Mu Z. Molecular signaling pathways in doxorubicin-induced nephrotoxicity and potential therapeutic agents. Int Immunopharmacol 2025; 144:113373. [PMID: 39566381 DOI: 10.1016/j.intimp.2024.113373] [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: 08/11/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 11/22/2024]
Abstract
Doxorubicin (DOX), an anthracycline chemotherapeutic agent, is extensively utilized in the clinical management of both solid and hematological malignancies. Nevertheless, the clinical application of this treatment is significantly limited by adverse reactions and toxicity that may arise during or after administration. Its cytotoxic effects are multifaceted, with cardiotoxicity being the most prevalent side effect. Furthermore, it has the potential to adversely affect other organs, including the brain, kidneys, liver, and so on. Notably, it has been reported that DOX may cause renal failure in patients and there is currently no effective treatment for DOX-induced kidney damage, which has raised a high concern about DOX-induced nephrotoxicity (DIN). Although the precise molecular mechanisms underlying DIN remain incompletely elucidated, prior research has indicated that reactive oxygen species (ROS) are pivotal in this process, triggering a cascade of detrimental pathways including apoptosis, inflammation, dysregulated autophagic flux, and fibrosis. In light of these mechanisms, decades of research have uncovered several DIN-associated signaling pathways and found multiple potential therapeutic agents targeting them. Thus, this review intends to delineate the DIN associated signaling pathways, including AMPK, JAKs/STATs, TRPC6/RhoA/ROCK1, YAP/TEAD, SIRTs, Wnt/β-catenin, TGF-β/Smad, MAPK, Nrf2/ARE, NF-κB, and PI3K/AKT, and to summarize their potential regulatory agents, which provide a reference for the development of novel medicines against DIN.
Collapse
Affiliation(s)
- Changxu Lu
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China; Department of Urology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jinwen Wei
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Can Gao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China.
| | - Dan Dong
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China.
| | - Zhongyi Mu
- Department of Urology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
43
|
Yang M, Zheng S, Zeng W, Zhan C, Yang Y, Yang F. Chronic exposure to low-dose MC-LR induces ileal inflammation in mice through the PI3K/AKT/mTOR pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025:1-9. [PMID: 39790025 DOI: 10.1080/15287394.2024.2441294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The global phenomenon of cyanobacterial bloom pollution is spreading globally due to climate change and eutrophication. It is well established that harmful cyanobacteria produce a wide range of toxins including microcystin-LR (MC-LR), a cyclic heptapeptide toxin known to damage various organs. The intestinal tract is the main site of MC-LR absorption and one of the targets susceptible to toxicity. Currently, studies on the enterotoxic effects of MC-LR predominantly focused on the colorectum, with limited investigations addressing the impact of microcystins on the small intestine. Therefore, the aim of our study was to examine the impact of chronic 9-month exposure of mice to low-dose 120 μg/L MC-LR in drinking water on ileal inflammation and potential mechanisms underlying these effects. Our findings showed that in mice chronically administered with low-dose MC-LR disorganized intestinal epithelial cells, lymphocytic infiltration and disturbed crypt arrangement were detected. The results of qPCR and Western blot demonstrated that, in comparison to control, the mRNA expression levels of pro-inflammatory factors IL-6, IL-17, IL-18, and IFN-γ were markedly elevated in the ileal tissue of mice treated with MC-LR, associated with significant increases in protein expression levels of p-PI3K, p-AKT, and p-mTOR. Taken together, evidence indicates that MC-LR induces ileal inflammation and histopathological damage involved activation of the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Mingjie Yang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuilin Zheng
- Changsha Center for Disease Control and Prevention, Changsha, China
| | - Wen Zeng
- The Department of Public Health, The Central Hospital of Shaoyang, Shaoyang, China
| | - Chunhua Zhan
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Yue Yang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- The Department of Public Health, The Central Hospital of Shaoyang, Shaoyang, China
| | - Fei Yang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Nuclear Medicine Department, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
44
|
Li Z, Zhang C, Wang L, Zhang Q, Dong Y, Sha X, Wang B, Zhu Z, Wang W, Wang Y, Zhou Y, Zhang Y. Chitooligosaccharides promote diabetic wound healing by mediating fibroblast proliferation and migration. Sci Rep 2025; 15:556. [PMID: 39747336 PMCID: PMC11697320 DOI: 10.1038/s41598-024-84398-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
Diabetic wounds are notoriously difficult to heal due to impaired cell repair mechanisms, reduced angiogenesis, and a heightened risk of infection. Fibroblasts play a vital role in wound healing by producing extracellular matrix (ECM) components and various growth factors, but their function is inhibited in diabetic wounds. Chitooligosaccharides (COS), intermediate products of chitosan degradation, have shown efficacy in promoting tissue repair, yet their role in diabetic wound healing remains underexplored. In a mouse model of diabetic wounds, COS treatment demonstrated substantial bioactivity in accelerating wound healing by enhancing fibroblast proliferation and migration. Additionally, COS increased collagen III deposition and angiogenesis at the wound sites. The COS also mitigated inflammatory responses by controlling leukocyte infiltration and bacterial infection. Mechanistically, COS regulated fibroblast activity via the PI3K/Akt signaling pathway, providing a novel bioactive material for chronic wound healing.
Collapse
Affiliation(s)
- Zihan Li
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
- Medical College, Nantong University, Nantong, People's Republic of China
| | - Chuwei Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
- Medical College, Nantong University, Nantong, People's Republic of China
| | - Lei Wang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
- Department of Burn and Plastic Surgery, Zhongda Hospital Affiliated Southeast University, Nanjing, People's Republic of China
| | - Qingrong Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military, Chongqing, People's Republic of China
| | - Yipeng Dong
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xinyu Sha
- Affiliated Hospital of Jiangnan University, Wuxi, People's Republic of China
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Bolin Wang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
- Medical College, Nantong University, Nantong, People's Republic of China
| | - Zhihan Zhu
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China
- Medical College, Nantong University, Nantong, People's Republic of China
| | | | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, People's Republic of China.
| | - Youlang Zhou
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China.
- The Hand Surgery Research Center, Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China.
| | - Yi Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People's Republic of China.
| |
Collapse
|
45
|
Ji J, Li X, Zhang R, Zhang J, Ren J, Du J, Su Z, Tian X, Wang Y, Xiang F, Li X. S100A4 exerts neuroprotective effects by attenuating blood-brain barrier disruption and oxidative stress via the PI3K/Akt/Nrf2 axis in ischemic stroke. Biochem Biophys Res Commun 2025; 742:151099. [PMID: 39657348 DOI: 10.1016/j.bbrc.2024.151099] [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: 11/21/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
Ischemic stroke is a major cause of disability and mortality worldwide, with oxidative stress and blood-brain barrier (BBB) injury playing crucial roles in its pathogenesis. Our RNA sequencing results revealed that S100 calcium-binding protein A4 (S100A4) is highly expressed in the middle cerebral artery occlusion (MCAO) mouse model. We analyzed S100A4 expression in ischemic stroke patients and in mice subjected to the MCAO model. Moreover, using adeno-associated virus (AAV)-mediated knockdown of S100A4 in mice, we evaluated its effects on neurological deficits, BBB integrity, and oxidative stress in MCAO mice. Bioinformatic analyses explored the potential downstream pathways of S100A4.S100A4 expression was significantly elevated in the serum of ischemic stroke patients and brain tissues of MCAO mice. AAV-mediated knockdown of S100A4 exacerbated neurological deficits, BBB disruption, and oxidative stress in MCAO mice. The upregulation of S100A4 mitigated these outcomes, which were facilitated through the stimulation of the PI3K/Akt/Nrf2 signaling cascade.Our results illustrate that S100A4 plays a protective role in preventing neuronal damage during ischemic stroke by reducing oxidative stress and preserving BBB integrity through the PI3K/Akt/Nrf2 pathway. This highlights its promise as a potential therapeutic approach for ischemic stroke.
Collapse
Affiliation(s)
- Jiyu Ji
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Xinxiang, Henan Province, 453100, China
| | - Xiao Li
- Department of Pharmacy, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Rong Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Xinxiang, Henan Province, 453100, China
| | - Jingjing Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Xinxiang, Henan Province, 453100, China
| | - Jing Ren
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Xinxiang, Henan Province, 453100, China
| | - Jia Du
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Xinxiang, Henan Province, 453100, China
| | - Zhou Su
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Xinxiang, Henan Province, 453100, China
| | - Xiaojun Tian
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Xinxiang, Henan Province, 453100, China
| | - Yumei Wang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Xinxiang, Henan Province, 453100, China
| | - Fang Xiang
- Department of Pharmacy, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China.
| | - Xiang Li
- Department of Pharmacy, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
46
|
Lu Y, Yuzhen H, Yi G, Lili W, Yan W, Weiwei T, Wanli L. Mechanism of Action of Tongjiang Mixture for Treating Reflux Esophagitis: A Study Using Serum Pharmacochemistry and Network Pharmacology. Adv Biol (Weinh) 2025; 9:e2400187. [PMID: 39601466 DOI: 10.1002/adbi.202400187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 09/13/2024] [Indexed: 11/29/2024]
Abstract
Tongjiang Mixture (TJM) is a traditional Chinese formula for treating reflux esophagitis (RE). Nevertheless, its active ingredients and potential pharmacological mechanisms are not yet clearly elucidated. This study will identify the active ingredients of TJM using serum pharmacochemistry and to elucidate the mechanism on RE through network pharmacology. The blood-borne ingredients of TJM are identified by the Ultra-performance liquid chromatography-quadrupole-time of flight-mass spectrometer. Subsequently, a "compound-target-disease" network is established and obtained core targets associated with TJM and RE. Then, the potential signaling pathways are forecasted through the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. Finally, the rat model of RE is established to verify the results predicted by network pharmacology through animal experiments. Fifteen blood-borne ingredients of TJM are identified, with eight active ingredients-namely Tangeretin, Tricin, Palmati, Berberine, Limonin, Evodiamine, Tetrahydropalmatine and Rutecarpine - making significant contributions to its efficacy. Moreover, TJM is predicted to act on 193 targets related to RE, involving AKT1, HSP90AA1, PIK3CA, and other targets, which enriches mainly in PI3K/AKT /NF-κB signaling. Additionally, TJM can alleviate inflammation of the esophageal mucosa, reduce pathological damage, and increase gastric pH. It can downregulate PI3K, AKT, and NF-κB mRNA transcription levels and reduce the protein expression of PI3K, AKT, and NF-κB. Furthermore, it can inhibit the overproduction of IL-6, TNF-α and IL-17. TJM can alleviate immune-inflammatory responses and ameliorate RE by restraining the PI3K/AKT pathway and its downstream NF-κB.
Collapse
Affiliation(s)
- Yang Lu
- Department of Gastroenterology, Nanjing Pukou District Hospital of Traditional Chinese Medicine, Nanjing, 211899, China
| | - Huang Yuzhen
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210012, China
| | - Gu Yi
- Department of Gastroenterology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Wu Lili
- School Of Integrative Medicine NJUCM, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | | | - Tao Weiwei
- School Of Integrative Medicine NJUCM, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Liu Wanli
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210012, China
| |
Collapse
|
47
|
Gomes-de-Pontes L, Barreiros LA, Gomes LN, Salgado RC, da Silva Napoleão SM, Soeiro-Pereira PV, Passos SD, Condino-Neto A. Congenital Zika Syndrome: Insights from Integrated Proteomic and Metabolomic Analysis. Biomolecules 2024; 15:32. [PMID: 39858427 PMCID: PMC11762526 DOI: 10.3390/biom15010032] [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: 08/07/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 01/27/2025] Open
Abstract
Background: In this study, we investigated the role of extracellular vesicles (EVs) in the pathogenesis of Congenital Zika Syndrome (CZS). Previous studies have highlighted the role of EVs in intercellular communication and the modulation of biological processes during viral infections, motivating our in-depth analysis. Our objective was to identify specific molecular signatures in the EVs of patients with CZS, focusing on their potential as biomarkers and on cellular pathways affected by the infection. Methods: We conducted advanced proteomic and metabolomic analyses using mass spectrometry for protein and metabolite identification. EVs were isolated from CZS patient samples and control groups using Izon qEV size-exclusion chromatography columns. Results: The analyzed EVs presented distinct molecular profiles in patients with CZS. Proteomic analysis revealed significant alterations in specific proteins, suggesting involvement in the PI3K-AKT-mTOR pathway, while metabolomics highlighted metabolites related to critical processes in Zika virus pathogenesis. These findings suggest a key role for the PI3K-AKT-mTOR pathway in regulating cellular processes during infection and indicate the involvement of EVs in intercellular communication. Additionally, the results identified potential biomarkers capable of aiding early diagnosis and assessing disease progression. Conclusions: This study demonstrates that EVs play a crucial role in intercellular communication during Zika virus infection. The identification of specific alterations in the PI3K-AKT-mTOR pathway highlights a possible therapeutic target, providing new opportunities for the development of more effective treatment strategies for CZS. Our findings significantly advance the understanding of CZS and underscore the need for further investigations using advanced techniques to validate and explore these potential molecular targets.
Collapse
Affiliation(s)
- Leticia Gomes-de-Pontes
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| | - Lucila Akune Barreiros
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| | - Lillian Nunes Gomes
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| | - Ranieri Coelho Salgado
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| | - Sarah Maria da Silva Napoleão
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| | | | - Saulo Duarte Passos
- Infectious Pediatric Laboratory, Medicine School of Jundiaí, Jundiaí 13202-550, SP, Brazil;
| | - Antonio Condino-Neto
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| |
Collapse
|
48
|
Gao C, Liu YJ, Yu J, Wang R, Shi JJ, Chen RY, Yang GJ, Chen J. Unraveling the Role of Ubiquitin-Conjugating Enzyme UBE2T in Tumorigenesis: A Comprehensive Review. Cells 2024; 14:15. [PMID: 39791716 PMCID: PMC11719737 DOI: 10.3390/cells14010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
Ubiquitin-conjugating enzyme E2 T (UBE2T) is a crucial E2 enzyme in the ubiquitin-proteasome system (UPS), playing a significant role in the ubiquitination of proteins and influencing a wide range of cellular processes, including proliferation, differentiation, apoptosis, invasion, and metabolism. Its overexpression has been implicated in various malignancies, such as lung adenocarcinoma, gastric cancer, pancreatic cancer, liver cancer, and ovarian cancer, where it correlates strongly with disease progression. UBE2T facilitates tumorigenesis and malignant behaviors by mediating essential functions such as DNA repair, apoptosis, cell cycle regulation, and the activation of oncogenic signaling pathways. High levels of UBE2T expression are associated with poor survival outcomes, highlighting its potential as a molecular biomarker for cancer prognosis. Increasing evidence suggests that UBE2T acts as an oncogene and could serve as a promising therapeutic target in cancer treatment. This review aims to provide a detailed overview of UBE2T's structure, functions, and molecular mechanisms involved in cancer progression as well as recent developments in UBE2T-targeted inhibitors. Such insights may pave the way for novel strategies in cancer diagnosis and treatment, enhancing our understanding of UBE2T's role in cancer biology and supporting the development of innovative therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| |
Collapse
|
49
|
Yue L, Li N, Ye X, Xiu Y, Wang B. Polymethoxylated flavones for modulating signaling pathways in inflammation. Int Immunopharmacol 2024; 143:113522. [PMID: 39515044 DOI: 10.1016/j.intimp.2024.113522] [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: 04/16/2024] [Revised: 09/18/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Aberrant signaling pathways play a crucial role in the pathogenesis of various diseases, including inflammatory disorders and autoimmune conditions. Polymethoxylated flavones (PMFs), a class of natural compounds found in citrus fruits, have obtained increasing attention for their potential therapeutic effects in modulating inflammatory responses. Although significant progress has been made in the pharmacological research of PMFs, the mechanisms by which they modulate signaling pathways to treat inflammation have not been systematically reviewed or analyzed. To address this gap in the literature, this review explores the mechanisms underlying the anti-inflammatory properties of PMFs and their prospects as drugs for treating inflammatory diseases. We discuss the molecular targets and signaling pathways through which PMFs exert their anti-inflammatory effects, including NF-κB pathway, PI3K/Akt pathway, MAPK pathway, Nrf2 pathway, and regulation of inflammatory cytokine production. Furthermore, we highlight preclinical studies evaluating the efficacy of PMFs in various inflammatory conditions, such as rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and osteoarthritis (OA). Despite promising findings, challenges remain in optimizing the pharmacokinetic properties and therapeutic efficacy of PMFs for clinical use. Future research directions include elucidating the structure-activity relationships of PMFs, developing novel delivery strategies, and conducting large-scale clinical trials to validate their efficacy and safety profiles. Overall, PMFs represent a promising class of natural compounds with potential applications as anti-inflammatory drugs, offering novel therapeutic opportunities for managing inflammatory diseases.
Collapse
Affiliation(s)
- Lixia Yue
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Ning Li
- Shenzhen Research Institute, the Hong Kong University of Science and Technology, Shenzhen 518054, China
| | - Xianglu Ye
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yanfeng Xiu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Bing Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
50
|
Mairuae N, Noisa P, Palachai N. Phytosome-Encapsulated 6-Gingerol- and 6-Shogaol-Enriched Extracts from Zingiber officinale Roscoe Protect Against Oxidative Stress-Induced Neurotoxicity. Molecules 2024; 29:6046. [PMID: 39770133 PMCID: PMC11677370 DOI: 10.3390/molecules29246046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
The rising prevalence of neurodegenerative disorders underscores the urgent need for effective interventions to prevent neuronal cell death. This study evaluates the neuroprotective potential of phytosome-encapsulated 6-gingerol- and 6-shogaol-enriched extracts from Zingiber officinale Roscoe (6GS), bioactive compounds renowned for their antioxidant and anti-inflammatory properties. The novel phytosome encapsulation technology employed enhances the bioavailability and stability of these compounds, offering superior therapeutic potential compared to conventional formulations. Additionally, the study investigates the role of the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)-signaling pathway, a key mediator of the neuroprotective effects of 6GS. Neurotoxicity was induced in SH-SY5Y cells (a human neuroblastoma cell line) using 200 μM of hydrogen peroxide (H2O2), following pretreatment with 6GS at concentrations of 15.625 and 31.25 μg/mL. Cell viability was assessed via the MTT assay alongside evaluations of reactive oxygen species (ROS), antioxidant enzyme activities (superoxide dismutase [SOD], catalase [CAT], glutathione peroxidase [GSH-Px]), oxidative stress markers (malondialdehyde [MDA]), and molecular mechanisms involving the PI3K/Akt pathway, apoptotic factors (B-cell lymphoma-2 [Bcl-2] and caspase-3), and inflammatory markers (tumor necrosis factor-alpha [TNF-α]). The results demonstrated that 6GS significantly improved cell viability, reduced ROS, MDA, TNF-α, and caspase-3 levels, and enhanced antioxidant enzyme activities. Furthermore, 6GS treatment upregulated PI3K, Akt, and Bcl-2 expression while suppressing caspase-3 activation. Activation of the PI3K/Akt pathway by 6GS led to phosphorylated Akt-mediated upregulation of Bcl-2, promoting neuronal survival and attenuating oxidative stress and inflammation. Moreover, Bcl-2 inhibited ROS generation, further mitigating neurotoxicity. These findings suggest that phytosome encapsulation enhances the bioavailability of 6GS, which through activation of the PI3K/Akt pathway, exhibits significant neuroprotective properties. Incorporating these compounds into functional foods or dietary supplements could offer a promising strategy for addressing oxidative stress and neuroinflammation associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Nootchanat Mairuae
- Biomedical Research Unit, Faculty of Medicine, Mahasarakham University, Mahasarakham 44000, Thailand;
| | - Parinya Noisa
- School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand;
| | - Nut Palachai
- Biomedical Research Unit, Faculty of Medicine, Mahasarakham University, Mahasarakham 44000, Thailand;
| |
Collapse
|