1
|
Yuan H, Liu J, Xu R, Yang K, Qu R, Liu S, Zhang Y, Xiang M. The spatiotemporal heterogeneity of reactive oxygen species in the malignant transformation of viral hepatitis to hepatocellular carcinoma: a new insight. Cell Mol Biol Lett 2025; 30:70. [PMID: 40517270 PMCID: PMC12167593 DOI: 10.1186/s11658-025-00745-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025] Open
Abstract
During the transformation of viral hepatitis into hepatocellular carcinoma (HCC), oxidative stress levels increase significantly, leading to tissue damage and chronic inflammation. HCC is characterized by spatiotemporal heterogeneity, which influences oxidative stress patterns, with reactive oxygen species (ROS) as the primary representative molecules. ROS serve not only as critical biomarkers of cancer but also as potential therapeutic targets for HCC, given that their increased levels can either promote or inhibit disease progression. In this review, we systematically examine the temporal heterogeneity of ROS, emphasizing its role in different stages of HCC progression caused by viral hepatitis and in influencing cell fate. We further explore ROS spatial heterogeneity at three levels: cellular, organelle, and biomolecular. Next, we comprehensively review clinical applications and potential therapies designed to selectively modulate ROS on the basis of its spatiotemporal heterogeneity. Finally, we discuss potential future applications of novel therapies that target ROS spatiotemporal heterogeneity to prevent and manage HCC onset and progression. In conclusion, this review enhances understanding of ROS in the progression of viral hepatitis to HCC and offers insights into developing new therapeutic targets and strategies centered on ROS heterogeneity.
Collapse
MESH Headings
- Humans
- Reactive Oxygen Species/metabolism
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/virology
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/virology
- Oxidative Stress
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Animals
- Hepatitis, Viral, Human/metabolism
- Hepatitis, Viral, Human/pathology
- Hepatitis, Viral, Human/complications
- Disease Progression
Collapse
Affiliation(s)
- Huimin Yuan
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Ruochen Xu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Keshan Yang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Ruiyang Qu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Shuai Liu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
| |
Collapse
|
2
|
Wu D, Fan F, He J, Wang L, Zhu C, Ji Y, Zhang H. Effects of polyunsaturated fatty acids on gastric cancer immunity and immunotherapy. Sci Rep 2025; 15:19891. [PMID: 40481119 PMCID: PMC12144108 DOI: 10.1038/s41598-025-97644-6] [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: 02/08/2025] [Accepted: 04/07/2025] [Indexed: 06/11/2025] Open
Abstract
Although immunotherapy has been predominant for advanced gastric cancer treatment, it still has limitations. Polyunsaturated fatty acids (PUFAs) are associated with inflammation while their roles in tumor immune microenvironment (TIME) are unclear. This study explores PUFAs' impacts on gastric cancer immunity and immunotherapy efficacy. Bioinformatics analysis was conducted to identify differential expressions of PUFAs metabolic genes and their immune correlations. Clinical data of advanced gastric cancer patients receiving immunotherapy at Zhongda Hospital (2020-2024), whose serum PUFAs were measured by mass spectrometry (MS), were collected and analyzed. Bioinformatics analysis revealed differential expression of half of PUFAs metabolic genes in gastric cancer. PUFAs metabolism towards Omega-3 (ω-3) tended to increase infiltration of active anti-tumor cells and up-regulate multiple immune checkpoints expressions, while metabolism towards Omega-6 (ω-6) led to opposite TIME outcomes. The clinical study demonstrated that lower serum ω-6/ω-3 ratio, arachidonic acid/eicosapentaenoic acid (AA/EPA) ratio, linoleic acid/alpha-linolenic acid (LA/ALA) ratio and higher EPA were associated with better six-month progression-free survival rate (6-month PFS) and one-year overall survival rate (1-year OS). This study deepens our understandings of TIME in gastric cancer. It clearly demonstrates that maintaining appropriate PUFAs ratios or values is promising in improving prognosis.
Collapse
Affiliation(s)
- Dikan Wu
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, China
| | - Fan Fan
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, China
| | - Junxian He
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, China
| | - Lian Wang
- Department of Oncology, Xuyi County People's Hospital, Huai'an, China
| | - Caiyun Zhu
- Department of Oncology, Xuyi County People's Hospital, Huai'an, China
| | - Yalin Ji
- Department of Oncology, Xuyi County People's Hospital, Huai'an, China
| | - Haijun Zhang
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, China.
| |
Collapse
|
3
|
Chen L, Guillot A, Tacke F. Reviewing the function of macrophages in liver disease. Expert Rev Gastroenterol Hepatol 2025; 19:621-637. [PMID: 40387555 DOI: 10.1080/17474124.2025.2508963] [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: 12/03/2024] [Revised: 04/10/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION The liver is a central metabolic organ, but is also hosting a unique immune microenvironment to sustain homeostasis and proper defense measures against injury threats in healthy individuals. Liver macrophages, mostly represented by the tissue-resident Kupffer cells and bone marrow- or monocyte-derived macrophages, are intricately involved in various aspects of liver homeostasis and disease, including tissue injury, inflammation, fibrogenesis and repair mechanisms. AREAS COVERED We review recent findings on defining the liver macrophage landscape and their functions in liver diseases with the aim of highlighting potential targets for therapeutic interventions. A comprehensive literature search in PubMed and Google Scholar was conducted to identify relevant literature up to date. EXPERT OPINION Liver macrophages orchestrate key homeostatic and pathogenic processes in the liver. Thus, targeting liver macrophages represents an attractive strategy for drug development, e.g. to ameliorate liver inflammation, steatohepatitis or fibrosis. However, translation from fundamental research to therapies remains challenging due to the versatile nature of the liver macrophage compartment. Recent and major technical advances such as single-cell and spatially-resolved omics approaches deepened our understanding of macrophage biology at a molecular level. Yet, further studies are needed to identify suitable, etiology- and stage-dependent strategies for the treatment of liver diseases.
Collapse
Affiliation(s)
- Lanlan Chen
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
4
|
Dong Z, Chen Z, Yu K, Zhao D, Jia J, Gao X, Wang D. Roles of plasma proteins in mediating the causal effect of the lipid species on gastric cancer: Insights from proteomic and two-step Mendelian randomization. Medicine (Baltimore) 2025; 104:e42485. [PMID: 40388730 PMCID: PMC12091653 DOI: 10.1097/md.0000000000042485] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 05/02/2025] [Indexed: 05/21/2025] Open
Abstract
The change of plasma lipid species has close contacts with gastric cancer (GC). However, the specific mechanism still needs to be explored further. We aim to utilize plasma proteins to decipher the association between lipid species and GC, and seek possible drug targets for GC. We performed a two-step Mendelian randomization (MR) analysis to investigate causal relationships among 179 lipid species, 4907 plasma proteins, and GC. Using summary-data-based MR and colocalization, we first examined protein-GC associations in discovery (N = 35,559) and validation (N = 54,219) cohorts. Subsequent MR analyses assessed lipid-GC and lipid-protein relationships, followed by mediation analysis using error propagation methods. Finally, macromolecular docking of prioritized proteins identified potential therapeutic ligands. Our MR analysis revealed causal relationships between 12 lipid species and GC, as well as 3 plasma proteins and GC. Importantly, mediation analysis demonstrated that CCDC80 protein mediates 2.90% (95% CI: 0.30-5.5%) of the protective effect of diacylglycerol (16:1_18:1) against GC. Based on these findings, we identified valproic acid as a promising therapeutic candidate targeting CCDC80 for GC treatment. Our study demonstrates that reduced CCDC80 expression mediates the tumor-promoting effects of diacylglycerol (16:1_18:1) in GC pathogenesis. Molecular docking confirms valproic acid binds stably to CCDC80, suggesting its therapeutic potential. These findings advance GC etiology understanding and provide a new drug development direction.
Collapse
Affiliation(s)
- Zhenhua Dong
- Gastric and Colorectal Surgery Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhiqing Chen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Kai Yu
- Urology Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dingliang Zhao
- Second Urology Department, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jianling Jia
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xulei Gao
- Second Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Daguang Wang
- Gastric and Colorectal Surgery Department, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Xu J, Liu X, Huang Z, Zhang Y, Li X, Cai D. NCAPG2 promotes the proliferation, metastasis and resistance to nab-paclitaxel in gastric adenocarcinoma cells. J Chemother 2025:1-10. [PMID: 40375564 DOI: 10.1080/1120009x.2025.2498230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 05/18/2025]
Abstract
Gastric adenocarcinoma is one of the most common cancers in the world. The purpose of this study was to investigate the role of non-smooth muscle cell aggregation protein II complex G2 subunit (NCAPG2) in gastric cancer cells. Cell growth, proliferation, migration, invasion and albumin-bound paclitaxel resistance were detected by cell counting kit-8, flow cytometry, Transwell chamber and nude mouse xenograft model. Western blot was used to detect the expression of NCAPG2, STAT3 and NF-κB signaling pathways. The expression of NCAPG2 was significantly up-regulated in gastric adenocarcinoma cells. Knockdown of NCAPG2 inhibited cell proliferation, migration and invasion, and inhibited tumor growth. In addition, knockdown of NCAPG2 reduced the resistance of gastric adenocarcinoma cells to albumin-bound paclitaxel and down-regulated STAT3 and NF-κB signaling pathways. In summary, NCAPG2 plays an important role in the malignant progression of gastric adenocarcinoma and is involved in the resistance to albumin-bound paclitaxel.
Collapse
Affiliation(s)
- Jia Xu
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoyuan Liu
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Zebo Huang
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Ying Zhang
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Xia Li
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Dongyan Cai
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
6
|
Geng Y, Yang L, Shao R, Xu T, Zhang L. RNA-binding protein gene NOP58 exhibits crucial prognostic and therapeutic value in Ewing sarcoma. Hereditas 2025; 162:76. [PMID: 40369667 PMCID: PMC12076867 DOI: 10.1186/s41065-025-00440-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/26/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Our aim was to identify crucial RNA-binding proteins (RBP) genes associated with Ewing sarcoma (EwS) in order to provide valuable insights into its mechanisms of tumorigenesis and to enhance therapeutic intervention. RESULTS Differential gene expression analysis identified candidate genes. Next, hub genes were generated by the results of protein-protein interaction (PPI) network, and univariate COX regression analysis. CIBERSORT was applied to analyze immune landscape. Furthermore, both in vitro and in vivo experiments were conducted to investigate the function of NOP58 in EwS. RESULTS A total of 179 RBP-related genes were significantly different in EwS tissues and normal controls. Among these, NOP58 ribonucleoprotein (NOP58) was considered as the hub gene, demonstrating significant prognostic value. Significantly, high NOP58 expression correlated with poor prognosis of EwS patients. Additionally, the levels of NOP58 were significantly up-regulated in EwS cells compared with human mesenchymal stem cells. Furthermore, knockdown of NOP58 notably inhibited the proliferation and migration of EwS cells. Moreover, NOP58 deficiency remarkably induced apoptosis and cell cycle arrest in EwS cells. In vivo studies on tumor-bearing mice demonstrated that NOP58 downregulation significantly inhibited tumor growth in EwS. CONCLUSION Collectively, downregulation of NOP58 could inhibit the proliferation and migration of EwS cells in vitro and reduce murine xenograft tumor growth in vivo. These findings identified NOP58 as a promising regulator of EwS tumorigenesis, suggesting it may serve as a potential therapeutic target for EwS treatment.
Collapse
Affiliation(s)
- Yannan Geng
- Department of the Sixth Spinal Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, No. 190 Jieyuan Road, Hongqiao District, Tianjin, 300121, China
| | - Lu Yang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Rui Shao
- Department of the Sixth Spinal Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, No. 190 Jieyuan Road, Hongqiao District, Tianjin, 300121, China
| | - Tiantong Xu
- Department of the Sixth Spinal Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, No. 190 Jieyuan Road, Hongqiao District, Tianjin, 300121, China.
| | - Lilong Zhang
- Department of the Sixth Spinal Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, No. 190 Jieyuan Road, Hongqiao District, Tianjin, 300121, China.
| |
Collapse
|
7
|
Chen C, Zheng M, Dong X, Zhang P, Bao Z, Cao Y, Liu Y, Yan J, Guo Y, Zeng X. Association of dietary inflammatory index with gynecological cancers in NHANES 2011-2018. Front Nutr 2025; 12:1560987. [PMID: 40421042 PMCID: PMC12104050 DOI: 10.3389/fnut.2025.1560987] [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: 01/15/2025] [Accepted: 04/25/2025] [Indexed: 05/28/2025] Open
Abstract
Objective This study aimed to analyze the association between the Dietary Inflammatory Index (DII) and the risk of gynecological cancers using data collected from the National Health and Nutrition Examination Survey (NHANES) between 2011 and 2018. Methods The data for this study were obtained from NHANES, conducted between 2011 and 2018, and included a total of 8,380 women. To examine the association between the Dietary Inflammatory Index and gynecological cancers, weighted multivariable logistic regression analyses were performed, using DII both as a continuous variable and as a categorical variable divided into tertiles. Subgroup analyses stratified by DII and gynecological cancer characteristics were conducted to further explore this association. Additionally, restricted cubic spline (RCS) analysis was applied to evaluate potential non-linear relationships between DII and gynecological cancer risk. Results Among the 8,380 women included in the analysis, the mean age was 47.02 (SD: 16.91) years, and 196 participants self-reported a diagnosis of gynecological cancer. In fully adjusted models, DII was significantly positively associated with the prevalence of gynecological cancer, whether analyzed as a continuous variable (OR = 1.15, 95% CI: 1.00-1.33, p = 0.046) or as a categorical variable (highest tertile compared to the lowest tertile: OR = 2.14, 95% CI: 1.14-4.04, p = 0.021, p for trend = 0.021). Restricted cubic spline analysis confirmed a linear relationship between DII and gynecological cancer risk (p for non-linear association = 0.1984). Subgroup analyses revealed a significant interaction effect with smoking status (p for interaction = 0.037). Conclusion Our findings suggest that higher DII scores are positively associated with an increased risk of gynecological cancer. These results contribute to the existing literature and underscore the need for further validation through larger prospective cohort studies.
Collapse
Affiliation(s)
- Chen Chen
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease’s Early Diagnosis, Zhengzhou, China
| | - Mengyu Zheng
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease’s Early Diagnosis, Zhengzhou, China
| | - Xing Dong
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease’s Early Diagnosis, Zhengzhou, China
| | - Pei Zhang
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease’s Early Diagnosis, Zhengzhou, China
| | - Zhuo Bao
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease’s Early Diagnosis, Zhengzhou, China
| | - Yushan Cao
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease’s Early Diagnosis, Zhengzhou, China
| | - Yixuan Liu
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease’s Early Diagnosis, Zhengzhou, China
| | - Jinxiang Yan
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease’s Early Diagnosis, Zhengzhou, China
| | - Yongzhen Guo
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease’s Early Diagnosis, Zhengzhou, China
| | - Xianxu Zeng
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease’s Early Diagnosis, Zhengzhou, China
| |
Collapse
|
8
|
Feng X, Qin Y, Feng Y, Zhuo Y. Research on the functions and potential mechanisms of STAT3 in chronic myelogenous leukemia. Discov Oncol 2025; 16:739. [PMID: 40353923 PMCID: PMC12069186 DOI: 10.1007/s12672-025-02492-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
OBJECTIVE To explore the bioinformatics characteristics and potential mechanisms of signal transducer and activator of transcription (STAT3) in chronic myelogenous leukemia (CML). METHODS Through the cancerSEA and CCLE databases, the expression of STAT3 in CML was verified and analyzed. Subsequently, K562 cells were treated with the STAT3 inhibitor Stattic. Western blotting, cell counting, and flow cytometry were utilized to observe its impact on the functions of K562 cells. Then, Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) were applied to deeply explore the regulatory mechanism of STAT3. The "LIMMA" software package was used to calculate STAT3-related differentially expressed genes (DEGs). Machine-earning methods were utilized to screen the STAT3-related hub genes. The "pROC" software package was employed to perform Receiver Operating Characteristic (ROC) curve analysis on the hub genes. The "corrplot" software package was used to conduct a correlation analysis of the hub genes. The "RMS" software package was applied to construct a nomogram of the hub genes. Based on the DisGENET database, a disease network of the hub genes was constructed, and the DGIdb database was used to construct a drug network of the hub genes. RESULTS In CML, the expression of STAT3 is upregulated compared to housekeeping genes. Among the 14 cell lines related to CML, STAT3 has the highest expression level in K562 cells. Stattic at a concentration of 5 μM can inhibit the proliferation of K562 cells, promote their apoptosis, and block the cell cycle at the S phase (P < 0.05). GSEA and GSVA indicates that amino acid metabolism, NOD-like receptor of STAT3. LASSO and SVM-RFE show that NCF4, PLAS1, IL7R, and TAGLN2 are hub differentially expressed genes (DEGs) related to STAT3. ROC and Nomogram indicate that the hub DEGs have high clinical diagnostic value. Correlation analysis shows that PLAS1 and NCF4 are negatively correlated, while PLAS1 and TAGLN2 are positively correlated. The construction of gene-disease networks reveals that these genes not only participate in the occurrence and development of CML but also jointly participate in multiple disease processes. The gene-drug network obtained 38 drugs targeting genes. CONCLUSION STAT3 might serve as a potential target for the treatment of CML. In CML, NCF4, PLAS1, IL7R, and TAGLN2 are hub genes associated with STAT3. These findings offer a fundamental theory for comprehending the pathogenesis of CML.
Collapse
Affiliation(s)
- Xiaoyun Feng
- Shizhen College of Guizhou University of Traditional Chinese Medicine, Guiyang, 550200, Guizhou, China.
| | - Yufeng Qin
- Department of Prosthodontics, Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Yulong Feng
- Shizhen College of Guizhou University of Traditional Chinese Medicine, Guiyang, 550200, Guizhou, China
| | - Yingquan Zhuo
- Department of Pediatric Surgery, The Afliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
9
|
Zhang D, Wang X, Xiao M, Ma S, Li S, Jia W. PLEKHA4 is transcriptionally regulated by HOXD9 and regulates glycolytic reprogramming and progression in glioblastoma via activation of the STAT3/SOCS-1 pathway. Oncogenesis 2025; 14:15. [PMID: 40346039 PMCID: PMC12064757 DOI: 10.1038/s41389-025-00559-0] [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: 07/29/2024] [Revised: 04/11/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
Recent studies have demonstrated that PLEKHA4 promotes tumor growth in some cancers, such as small-cell lung cancer, melanoma, and hepatic carcinomas; however, the underlying mechanism in glioblastoma remains ambiguous. Bioinformatic was used to analysis PLEKHA4 expression. In vitro and in vivo experiments were conducted to detect the effect of PLEKHA4 on glioblastoma cell glycolytic reprogramming and progression. GSEA was used to analyze the signal pathways related to PLEKHA4. Pharmacological methods further validated the role of activation pathways. We evaluated the effects of PLEKHA4 knockdown combined with temozolomide (TMZ) on glioblastoma cell proliferation and apoptosis in vitro and in vivo. We observed an overexpression of PLEKHA4 in GBM cell lines, resulting in enhanced cell proliferation, inhibited apoptosis, and promoted glycolysis. Mechanistically, our study demonstrated that PLEKHA4 mediates cell proliferation, apoptosis, and glycolysis via the STAT3/SOCS1 signaling pathway. Additionally, HOXD9 was predicted using Jasper, which is a transcription factor that binds to the PLEKHA4 promoter region. Knocking down PLEKHA4 combined with TMZ inhibited cell proliferation and promoted cell apoptosis in vitro and in vivo. Our results indicated that HOXD9-medicated PLEKHA4 regulates glioblastoma cell proliferation and glycolysis via activation of the STAT3/SOCS1 pathway.
Collapse
Affiliation(s)
- Dainan Zhang
- Department of Neurosurgery, Beijing Tian Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xiaoyin Wang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Meng Xiao
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
- Graduate School of Beijing University of Chinese Medical, Beijng, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tian Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shaomin Li
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Wang Jia
- Department of Neurosurgery, Beijing Tian Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
10
|
Zhao Z, Xiang L, Hong JS, Wang Y, Feng J. Mechanisms of Acetate in Alleviating SETDB1-Linked Neuroinflammation and Cognitive Impairment in a Mouse Model of OSA. J Inflamm Res 2025; 18:5931-5950. [PMID: 40357375 PMCID: PMC12067661 DOI: 10.2147/jir.s510690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Background Microglia-mediated neuroinflammation is crucial for obstructive sleep apnea (OSA)-induced cognitive impairment. We aimed to investigate roles of acetate (ACE) and SET domain bifurcated histone lysine methyltransferase 1 (SETDB1) in neuroinflammation of OSA. Methods After C57BL/6J mice were exposed to OSA-associated intermittent hypoxia (IH) or normoxia for four weeks, the composition of the gut microbiota (GM) and the levels of serum short-chain fatty acids (SCFAs) were measured by 16S rRNA and GC-MS methods, respectively. To assess the effect of ACE on IH mice, glyceryl triacetate (GTA) was gavaged in IH-exposed mice and the cognitive function, microglial activation, and hippocampal neuronal death were examined. Moreover, ACE-treated BV2 microglia cells were also utilized for further mechanistic studies. Results IH disrupts the gut microbiome, reduces microbiota-SCFAs, and impairs cognitive function. Gavage with GTA significantly mitigated these cognitive deficits. Following IH exposure, we observed substantial increases in SETDB1 both in vivo and in vitro, along with elevated levels of histone H3 lysine 9 trimethylation (H3K9me3). Genetic or pharmacological inhibition of SETDB1 in microglia led to decreased induction of proinflammatory factors, as well as reduced reactive oxygen species (ROS) generation. Mechanistically, SETDB1 was found to upregulate the transcription factors p-signal transducer and activator of transcription 3 (p-STAT3) and p-NF-κB. In vitro, ACE supplementation effectively repressed high SETDB1 and H3K9me3 levels, thereby inhibiting microglial pro-inflammatory responses induced by IH. In vivo, ACE supplementation significantly reduced hippocampal levels of p-STAT3, p-NF-κB, and pro-inflammatory cytokines while also protecting neuronal integrity. Conclusion This study provides the first evidence that H3K9 methyltransferase SETDB1 promotes microglial pro-inflammatory response distinct from its previously shown role in macrophages. Our findings also identify ACE supplementation as a promising dietary intervention for OSA-related cognitive impairment with SETDB1 serving as both a mechanistic biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Zhan Zhao
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| | - Li Xiang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Yubao Wang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| |
Collapse
|
11
|
Fu S, Lv R, Wang L, Wang Z, Wang F, Gao H, Zhao W, Huang X, Li X, Wang Y. Bone mesenchymal stem cells based on matric hydrogels attenuate intervertebral disc degeneration by suppressing oxidative stress-induced ferroptosis. Sci Rep 2025; 15:15378. [PMID: 40316606 PMCID: PMC12048664 DOI: 10.1038/s41598-025-00278-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 04/28/2025] [Indexed: 05/04/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) and its attendant lower back pain are a major medical challenge. Ferroptosis has become a new target for the treatment of IVDD. Mesenchymal stem cells (MSCs) are a promising regenerative therapy for IVDD. Hydrogel is usually used as a delivery carrier for MSCs. This study investigated the effect of bone mesenchymal stem cells (BMSCs) in IVDD by magnetic resonance imaging (MRI) and hematoxylin and eosin (HE) staining analysis using a rat-punctured IVDD model. A vitro model of tert-butyl hydroperoxide (TBHP)-induced oxidative stress injury in annulus fibrosus cells (AFCs) was used to explore the underlying molecular mechanisms. Cell viability was detected by cell counting kit-8 assay. Ferroptosis was assessed by measuring the levels of LDH, Fe2+, glutathione, lipid reactive oxygen species, and malondialdehyde. The underlying mechanism was investigated by western blot and phosphor-kinase array. Results suggested that BMSCs inhibited TBHP-induced ferroptosis and the phosphorylated levels of STAT3 in AFCs. The activation of STAT3 (colivelin, a specific agonist for STAT3) reversed the effects on the ferroptosis of BMSCs. Additionally, BMSCs alleviated IVDD progression based on matrix hydrogels, while colivelin abolished the protective effects of BMSCs-encapsulated hydrogels on IVDD. In short, BMSCs inhibited oxidative stress-induced AFCs ferroptosis, thereby alleviating IVDD, which is associated with inhibited STAT3 activation. This study demonstrated the possible underlying mechanism by which BMSCs mitigate IVDD and may provide a new therapeutic idea for IVDD.
Collapse
Affiliation(s)
- Song Fu
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, No.1, Fengshan Road, Wendeng District, Weihai, 264400, Shandong Province, China
| | - Renhua Lv
- Department of Neurology, Weihai Central Hospital, Weihai, Shandong Province, China
| | - Longqiang Wang
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, No.1, Fengshan Road, Wendeng District, Weihai, 264400, Shandong Province, China
| | - Zhenyu Wang
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, No.1, Fengshan Road, Wendeng District, Weihai, 264400, Shandong Province, China
| | - Fengming Wang
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, No.1, Fengshan Road, Wendeng District, Weihai, 264400, Shandong Province, China
| | - Hao Gao
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, No.1, Fengshan Road, Wendeng District, Weihai, 264400, Shandong Province, China
| | - Wei Zhao
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, No.1, Fengshan Road, Wendeng District, Weihai, 264400, Shandong Province, China
| | - Xiaoling Huang
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, No.1, Fengshan Road, Wendeng District, Weihai, 264400, Shandong Province, China
| | - Xiaojun Li
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, No.1, Fengshan Road, Wendeng District, Weihai, 264400, Shandong Province, China
| | - Yanan Wang
- Department of Minimally Invasive Spine Surgery, Shandong Wendeng Orthopedic Hospital, No.1, Fengshan Road, Wendeng District, Weihai, 264400, Shandong Province, China.
| |
Collapse
|
12
|
Dai YW, Wu ZX, Cheng Y, Wu HD, Chen JW, Lv LX, Wang ZQ, Li HF, Yan CZ, Bao JX, Liu CH, Dai XX. Formosanin C inhibits triple-negative breast cancer progression by suppressing the phosphorylation of STAT3 and the polarization of M2 macrophages. Breast Cancer Res Treat 2025; 211:71-89. [PMID: 39953272 DOI: 10.1007/s10549-025-07623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 01/20/2025] [Indexed: 02/17/2025]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC), is a highly aggressive tumor. Formosanin C (FC) is a diosgenin with immunomodulatory and antitumor properties, the precise mechanism through which it is against TNBC remains uncertain. OBJECTIVE Clarifying the mechanism of FC against TNBC. MATERIALS AND METHODS The impact of FC on two TNBC cell lines for 24 h was investigated through various techniques including the CCK8 assay, flow cytometry, transwell assay, scratch tests, immunoblot assay, and immunofluorescence. To elucidate the mechanism behind the anti-TNBC effect of FC, MDA-MB-231 cells were subjected to STAT3 overexpression. Moreover, the in vivo efficacy of FC was examined using a xenograft nude mice (BALB/C). Mice were divided into the control group (equal amount of PBS), the napabucasin group (5 mg/kg) and the FC groups (1 mg/kg, 2 mg/kg). The study duration was 30 days. RESULTS FC exhibited inhibitory effects against MDA-MB-231 and Hs578T cells. FC can decrease the migratory capacity of TNBC cells by inhibiting epithelial-mesenchymal transition (EMT). Meanwhile, we demonstrated that the inhibition of phosphorylation of STAT3 (Y705) is the crucial mechanism of FC against TNBC. Moreover, FC also hindered the polarization of macrophage M2. DISCUSSION AND CONCLUSION This study is the first to show that FC restrains the EMT of TNBC cells by obstructing the STAT3 pathway and hinders the M2 polarization of macrophages and immune evasion. Therefore, FC holds the possibility of being utilized as a therapeutic remedy for TNBC.
Collapse
Affiliation(s)
- Yin-Wei Dai
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhi-Xuan Wu
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yao Cheng
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hao-Dong Wu
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia-Wei Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lin-Xi Lv
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zi-Qiong Wang
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hong-Feng Li
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cong-Zhi Yan
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing-Xia Bao
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cong-Hui Liu
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuan-Xuan Dai
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
13
|
Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ballal S, Sharma R, Debnath S, Sinha A, Rekha A, Khan NH, Alrashoud MM, Kamal M, Imran M. Pathological interplay of NF-κB and M1 macrophages in chronic inflammatory lung diseases. Pathol Res Pract 2025; 269:155903. [PMID: 40081284 DOI: 10.1016/j.prp.2025.155903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/25/2024] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
Inflammatory lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), and pulmonary fibrosis depend on the pathology of the nuclear factor kappa B (NF-κB) signalling pathway and M1 macrophage polarization. This review discusses the intimate molecular interactions and processes that modulate NF-κB's promotion of M1 macrophages and chronic inflammation/tissue damage within the confines of this review. NF-κB activation in macrophages produces pro-inflammatory mediators (cytokines - TNFα, IL6, IL1β, and reactive oxygen species (ROS), further increasing airway remodeling and fibrosis. MAPK, JAK-STAT, and PI3K-Akt signalling systems cross-talked with the pathway, amplifying its effect on lung disease progression. Therapeutic strategies focused on inhibiting this axis, including inhibition of NF-κB and small molecule/modulation of macrophage polarization, represent potential ways to attenuate inflammation and promote tissue repair. The potential of precision medicine is illustrated by natural compounds such as curcumin and resveratrol and innovative RNA-based and nanoparticle delivery systems. Despite these challenges, specificity, minimizing systemic side effects, and optimized delivery methods remain difficult. To develop targeted therapies, more research must be conducted to refine targeted approaches, including immune profiling and single-cell analysis. This review aims to advance the management of hard-to-treat inflammatory lung diseases by addressing these complexities.
Collapse
Affiliation(s)
- Fadiyah Jadid Alanazi
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Al Jouf City 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rajesh Sharma
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Sourav Debnath
- Chandigarh pharmacy college, Chandigarh Group of colleges, Jhanjeri, Mohali 140307, Punjab, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - A Rekha
- Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | | | - Muhanad Mubarak Alrashoud
- Department of Inpatient Pharmacy, Dr. Sulaiman Alhabib Hospital, Alhamra Branch, Riyadh 13333, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohd Imran
- Center for Health Research, Northern Border University, Arar, Saudi Arabia; Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| |
Collapse
|
14
|
Hai JJ, Liang W, Sun D, Yin P, Han B, Qu X. Rutin Attenuates Distraction Spinal Cord Injury by Inhibiting Microglial Inflammation Through Downregulation of P38 MAPK/NF-κB/STAT3 Pathway. Mol Neurobiol 2025; 62:6027-6040. [PMID: 39699845 DOI: 10.1007/s12035-024-04659-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Abstract
Distraction spinal cord injury (DSCI) is a severe complication following scoliosis correction surgery, for which there are currently no effective clinical treatments. This study aims to evaluate the inhibitory effects of rutin, a natural product, on inflammation in DSCI and to investigate the underlying mechanisms. In vitro, microglial cells were exposed directly to rutin to assess its ability to inhibit lipopolysaccharide (LPS)-induced inflammation. In rats with DSCI, the inhibitory effect of rutin on DSCI was evaluated using behavioral tests. mRNA sequencing was performed on spinal cord tissues to elucidate the mechanism of rutin's action. Rutin significantly suppressed the LPS-induced increase in inflammatory factors in microglial cells. In DSCI rats treated with rutin, scores in the Basso-Beattie-Bresnahan (BBB) were significantly improved. The mechanism of rutin's action was found to be related to its ability to reduce inflammatory infiltration in spinal cord tissue, protecting neurons from apoptosis and microstructural demyelination. Through assays of transcriptomic differentially expressed genes (DEGs), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and RT-qPCR validation of the top DEGs, MAPK13 (also known as P38 MAPK) was finally identified as the key target gene in promoting DSCI development. Further molecular docking analysis indicated an interaction between rutin and P38 MAPK, supporting the rutin's action and the underlying mechanism in anti-inflammation. In conclusion, rutin effectively inhibited the development of DSCI in rats. The mechanism of rutin's action was associated with its activity in blocking the P38 MAPK/NF-κB/STAT3 pathway in the microglial cells of spinal cord. Rutin could be developed as a potential anti-DSCI drug for clinical applications.
Collapse
Affiliation(s)
- Junrui Jonathan Hai
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Fengtai District, Beijing, 100069, China
- Princeton International School of Mathematics and Science, Princeton, NJ, 08540, USA
| | - Weishi Liang
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Fengtai District, Beijing, 100069, China
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Chaoyang District, Beijing, 100020, China
| | - Duan Sun
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Fengtai District, Beijing, 100069, China
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Chaoyang District, Beijing, 100020, China
| | - Peng Yin
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Fengtai District, Beijing, 100069, China
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Chaoyang District, Beijing, 100020, China
| | - Bo Han
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Fengtai District, Beijing, 100069, China.
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Chaoyang District, Beijing, 100020, China.
- Beijing Jishuitan Hospital, Capital Medical University, 31 Xinjiekou East Street, Xicheng District, Beijing, 100035, China.
| | - Xianjun Qu
- Joint Laboratory for Research & Treatment of Spinal Cord Injury in Spinal Deformity, Laboratory for Clinical Medicine, Capital Medical University, Fengtai District, Beijing, 100069, China.
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Fengtai District, Beijing, 100069, China.
| |
Collapse
|
15
|
Sheikh SM, Staab J, Bleyer M, Ivetic A, Lühder F, Wirths O, Meyer T. N-terminal truncation of STAT1 transcription factor causes CD3- and CD20-negative non-Hodgkin lymphoma through upregulation of STAT3-mediated oncogenic functions. Cell Commun Signal 2025; 23:201. [PMID: 40287766 PMCID: PMC12034123 DOI: 10.1186/s12964-025-02183-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/29/2025] [Indexed: 04/29/2025] Open
Abstract
The cytokine-driven transcription factor STAT1 (signal transducer and activator of transcription 1) executes anti-microbial and pro-apoptotic functions, and loss-of-function mutations are associated with increased susceptibility to various infections and the development of tumors. A targeted mutation in mice expressing an N-terminally truncated STAT1 protein (STAT1-ΔN) typically develops splenomegaly in animals older than 6 months due to the formation of splenic non-Hodgkin lymphomas. The expression of the STAT1-ΔN variant resulted in the disruption of normal spleen architecture by malignant CD3- and CD20-negative tumor cells, which stained positively for both tyrosine-phosphorylated STAT1 and STAT3. Immunoblotting of lysates from isolated tumor cells revealed the cytokine-independent hyperphosphorylation of both STAT proteins, whereas the expression level of NF-κB was significantly reduced. Gel-shift assays showed that the DNA-binding activity of STAT1-ΔN was increased compared to the wild-type protein. This elevated level of tyrosine-phosphorylated STAT1-ΔN did not further increase upon stimulation of isolated tumor cells with either interferon-γ (IFNγ), lipopolysaccharide (LPS), or the combination of both. Since the truncation mutant was unable to accumulate in the nucleus upon cytokine stimulation, real-time PCR data from tumor tissue as well as from isolated, IFNγ/LPS-treated lymphoma cells demonstrated significantly reduced STAT1-regulated target gene expression despite its observed hyperphosphorylation. The nuclear import defect of tyrosine-phosphorylated STAT1-ΔN was associated with an elevated tyrosine-phosphorylation level of its antagonistic homolog STAT3, which is a known oncogene. These data demonstrate that the lack of STAT1 nuclear accumulation interferes with the functional balance between the two STAT proteins and, thereby, promotes the formation of phospho-STAT3-expressing CD3-/- CD20-/- non-Hodgkin lymphomas in the spleens of the diseased animals.
Collapse
Affiliation(s)
- Sana Mumtaz Sheikh
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Institute of Pathology, University Medical Center Düsseldorf, Düsseldorf, Germany
| | - Julia Staab
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Martina Bleyer
- German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Aleksandar Ivetic
- School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Fred Lühder
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Oliver Wirths
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas Meyer
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
16
|
Barman P, Hazarika S, Roy K, Rawal RK, Konwar R. Phytochemical analysis of leaf extract of Piper nigrum and investigation of its biological activities. Inflammopharmacology 2025:10.1007/s10787-025-01701-5. [PMID: 40251438 DOI: 10.1007/s10787-025-01701-5] [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: 11/07/2024] [Accepted: 01/31/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND This study investigates the phytoconstituents of the less explored leaf of Piper nigrum, a common ethnomedicinal plant as an alternate source for multiple bioactivities. METHODS Hydro-ethanolic (1:4) extract of Piper nigrum leaves (PNLE) prepared and profiled using liquid chromatography and mass spectrometry for identification of phytomolecules. Anti-oxidant activity, intracellular reactive oxygen species (ROS) expression, phagocytosis activity, and cytokine expression were estimated using cell-free and cell-based assays. Anti-cancer activity was determined with cancer cell viability, migration inhibition and colony-formation assay. Apoptosis and membrane depolarization assay were done using fluorescent microscopic staining methods while network pharmacology, and molecular docking analysis were done using open source and online tools. RESULTS Major phytomolecules identified in PNLE were pentanamide N,N-didecyl, piperettine, curcumin, myristicin, pipernonaline, sesamin, and lupenone. PNLE exhibited anti-bacterial activity with higher activity against Gram-positive bacteria, Staphylococcus aureus. PNLE also showed anti-oxidant and anti-inflammatory activity through neutralization of free radicals; inhibition of intracellular ROS generation; inhibition of phagocytosis and reduction of cytokine (IL-6 and TNF-α) levels. PNLE showed anti-proliferative activity against human breast cancer cells (MDA-MB-231), rat mammary tumor cells (LA7), and mouse melanoma cells (B16-F10) with highest activity against MDA-MB-231 cells. The extract did not inhibit human kidney cells (HEK-293). Further, PNLE treatment significantly inhibited cell migration and colony formation of MDA-MB-231 cells. Fluorescent staining techniques confirmed induction of apoptosis in cancer cells by PNLE. Further, network pharmacology and molecular docking studies revealed that the identified PNLE phytomolecules share 97 targets of out of potential breast cancer and inflammation-related target genes with four best common target proteins among the top hub genes and sesamin showed the highest binding affinity with these important cellular targets. CONCLUSIONS Overall, the phytochemical profile of PNLE showed clear presence of important phytomolecules and their association with critical human cellular mechanistic pathways responsible for exhibited bioactivities. This study further establishes the leaf of P. nigrum as an additional anatomical plant part with potent medicinal properties and as a potential renewable source for bioactive phyomolecules.
Collapse
Affiliation(s)
- Pankaj Barman
- Centre for Preclinical Studies (CPS), Biological Science and Technology Division (BSTD), CSIR-North East Institute of Science and Technology (NEIST), Jorhat, Assam, 785006, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Srija Hazarika
- Natural Product Chemistry Group, Chemical Science and Technology Division (CSTD), CSIR-North East Institute of Science and Technology, Jorhat, 785006, India
| | - Kallol Roy
- Centre for Preclinical Studies (CPS), Biological Science and Technology Division (BSTD), CSIR-North East Institute of Science and Technology (NEIST), Jorhat, Assam, 785006, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ravindra K Rawal
- Natural Product Chemistry Group, Chemical Science and Technology Division (CSTD), CSIR-North East Institute of Science and Technology, Jorhat, 785006, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rituraj Konwar
- Centre for Preclinical Studies (CPS), Biological Science and Technology Division (BSTD), CSIR-North East Institute of Science and Technology (NEIST), Jorhat, Assam, 785006, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
17
|
Luo W, Zhou J, Liang F, Chou X, Peng Z, Tan W, Yu Z, Wan H. The GPR4 antagonist NE 52-QQ57 prevents ox-LDL-induced cellular senescence by promoting the expression of SIRT1. Genes Genomics 2025:10.1007/s13258-024-01610-x. [PMID: 40208484 DOI: 10.1007/s13258-024-01610-x] [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: 05/14/2024] [Accepted: 09/12/2024] [Indexed: 04/11/2025]
Abstract
BACKGROUND Cell senescence-associated endothelia dysfunction is a vital point in the pathological progression of atherosclerosis (AS). G-protein coupled receptor 4 (GPR4) is a proton-sensing receptor involved in developing endothelial dysfunction. OBJECTIVE In this study, we investigated the protective role of NE 52-QQ57, a GPR4 inhibitor in endothelial cell senescence induced using an oxidized low-density lipoprotein (ox-LDL). We also unravel the underlying molecular mechanism of NE 52-QQ57 as a therapeutic agent. METHODS Endothelial cell senescence model was established using human aortic endothelial cells (HAECs) stimulated with ox-LDL. The expression levels of GPR4, p53, p16, and sirtuin1 (SIRT1) were evaluated using real-time PCR and western blot assays. ROS production was determined using dihydroethidium (DHE) staining. Further, interleukin-6 (IL-6) and monocyte chemotactic protein 1 (MCP-1) secretion and expression were determined using ELISA and real-time PCR analysis, respectively. Finally, β-galactosidase (SA-β-Gal) staining associated with cell senescence, telomerase activity, and cell cycle assay were used to determine the state of cell senescence. RESULTS Firstly, GPR4 was found to be upregulated in the ox-LDL-stimulated HAECs. We also identified elevated ROS, IL-6, and MCP-1 levels induced by ox-LDL and significantly abrogated by NE 52-QQ57 treatment. Second, a reversal in SA-β-Gal activity, telomerase activity, and G0/G1 proportion, with an upregulation in p53 and p16 expressions was observed on NE 52-QQ57 treatment in the ox-LDL induced model. Lastly, the decreased expression level of SIRT1 was extremely elevated by NE 52-QQ57. Notably, the inhibitory effect of NE 52-QQ57 against ox-LDL-induced cell senescence was abolished by the SIRT1 inhibitor EX-527. CONCLUSION The GPR4 antagonist NE 52-QQ57 might prevent cellular senescence by promoting the expression of SIRT1.
Collapse
Affiliation(s)
- Wei Luo
- Department of Cardiology, The First Affiliated Hospital of University of South China, No. 69, Chuanshan Road, Shigu District, Hengyang, 421001, Hunan, China
| | - Jiming Zhou
- Department of Cardiology, The First Affiliated Hospital of University of South China, No. 69, Chuanshan Road, Shigu District, Hengyang, 421001, Hunan, China
| | - Feng Liang
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Xianghui Chou
- Department of Cardiology, The First Affiliated Hospital of University of South China, No. 69, Chuanshan Road, Shigu District, Hengyang, 421001, Hunan, China
| | - Zhengliang Peng
- Department of Emergency, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Weihua Tan
- Department of Emergency, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Ziying Yu
- Department of Emergency, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Huan Wan
- Department of Cardiology, The First Affiliated Hospital of University of South China, No. 69, Chuanshan Road, Shigu District, Hengyang, 421001, Hunan, China.
| |
Collapse
|
18
|
Lee JJ, Yang L, Kotzin JJ, Ahimovic D, Bale MJ, Nigrovic PA, Josefowicz SZ, Mathis D, Benoist C. Early transcriptional effects of inflammatory cytokines reveal highly redundant cytokine networks. J Exp Med 2025; 222:e20241207. [PMID: 39873673 PMCID: PMC11865922 DOI: 10.1084/jem.20241207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/25/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
Inflammatory cytokines are fundamental mediators of the organismal response to injury, infection, or other harmful stimuli. To elucidate the early and mostly direct transcriptional signatures of inflammatory cytokines, we profiled all immunologic cell types by RNAseq after systemic exposure to IL1β, IL6, and TNFα. Our results revealed a significant overlap in the responses, with broad divergence between myeloid and lymphoid cells, but with very few cell-type-specific responses. Pathway and motif analysis identified several main controllers (NF-κB, IRF8, and PU.1), but the largest portion of the response appears to be mediated by MYC, which was also implicated in the response to γc cytokines. Indeed, inflammatory and γc cytokines elicited surprisingly similar responses (∼50% overlap in NK cells). Significant overlap with interferon-induced responses was observed, paradoxically in lymphoid but not myeloid cell types. These results point to a highly redundant cytokine network, with intertwined effects between disparate cytokines and cell types.
Collapse
Affiliation(s)
- Juliana J. Lee
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Liang Yang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Jonathan J. Kotzin
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dughan Ahimovic
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Michael J. Bale
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Peter A. Nigrovic
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven Z. Josefowicz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
19
|
Li X, Guo Y, Deng X, Jiao Y, Hao H, Dong Q, Sun H, Han S. Taraxacum mongolicum Hand.-Mazz. extract disrupts the interaction between triple-negative breast cancer cells and tumor-associated macrophages by inhibiting RAC2/NF-κB p65/p38 MAPK pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119757. [PMID: 40199407 DOI: 10.1016/j.jep.2025.119757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/31/2025] [Accepted: 04/05/2025] [Indexed: 04/10/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Taraxacum mongolicum Hand.-Mazz., generally known as dandelion, is a herb renowned for its pharmacological properties, including detoxifying and anti-inflammatory effects. Historically, this herb has been extensively utilized in the treatment of breast diseases. Recent studies have demonstrated that dandelion exhibits inhibitory properties against triple-negative breast cancer (TNBC) and modulates the tumor-associated macrophages (TAMs) microenvironment. However, the primary pharmacological mechanisms remain to be completely revealed. AIM OF THE STUDY This study is focused on examining the mechanism by which dandelion extract regulates the communication between TNBC and TAMs through an integrative approach of network-based pharmacology and experimental verification. MATERIALS AND METHODS A three-dimensional (3D) co-culture cell model was employed to visualize the impact of dandelion extract on the crosstalk between TAMs and TNBC. To shed light on the crucial mechanisms of dandelion inhibitory effects on TNBC, a network pharmacology analysis was undertaken. Transwell assays were utilized to assess cell capabilities to migrate and infiltrate. Small interfering RNA (siRNA) and an overexpression plasmid targeting Ras-related C3 botulinum toxin substrate 2 (RAC2) were applied to knock down or upregulate the expression levels of RAC2. The altered expression levels of associated molecules were evaluated using quantitative real-time PCR, Western blotting, and immunohistochemistry. RESULTS The results from 3D co-culture model demonstrated that dandelion extract significantly hindered the consolidating strength between TNBC cells and TAMs. The extract effectively suppressed TAM-induced epithelial-mesenchymal transition (EMT) in TNBC cells and inhibited the recruitment of TAMs and M2 polarization mediated by TNBC cells. Network pharmacology analysis predicted that dandelion extract attenuates the inflammatory response in TNBC through NF-κB p65/p38 MAPK. Notably, dandelion extract reduced the levels of NF-κB p65/p38 MAPK-related cytokines, including TNF-α, IL-1β, and IL-6 in TNBC cells, while increasing them in TAMs. Overexpression of RAC2 in TNBC cells not only augmented their proliferation, migration, invasion, and EMT processes but also facilitated increased recruitment and M2 polarization of TAMs. TAMs were observed to promote lung metastasis, whereas dandelion extract significantly inhibited lung metastasis and EMT in 231 xenografts. Mechanically, dandelion extract significantly mitigated the RAC2/NF-κB p65/p38 MAPK-mediated inflammatory response both in TNBC cells and 231 xenografts, thereby disrupting the crosstalk between TNBC cells and TAMs. CONCLUSION Dandelion extract inhibits the crosstalk between TNBC and TAMs through RAC2/NF-κB p65/p38 MAPK inflammatory pathway, thereby suppressing lung metastasis in TNBC. This study revealed dandelion extract exerts a bidirectional regulatory effect on inflammation in modulating the interaction between TNBC and TAMs, offering a promising therapeutic insight for TNBC treatment.
Collapse
Affiliation(s)
- Xinrui Li
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, PR China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, 100142, PR China
| | - Yang Guo
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, PR China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, 100142, PR China
| | - Xinxin Deng
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, PR China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, 100142, PR China
| | - Yanna Jiao
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, PR China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, 100142, PR China
| | - Huifeng Hao
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, PR China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, 100142, PR China
| | - Qingqing Dong
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, PR China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, 100142, PR China
| | - Hong Sun
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, PR China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, 100142, PR China.
| | - Shuyan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, PR China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, 100142, PR China.
| |
Collapse
|
20
|
Yang TT, Lan CCE. Photocarcinogenesis of the skin: Current status and future trends. Kaohsiung J Med Sci 2025; 41:e12946. [PMID: 39907400 DOI: 10.1002/kjm2.12946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025] Open
Abstract
Solar radiation is essential for life on Earth but is also a major contributor to skin carcinogenesis. Solar radiation, particularly ultraviolet (UV) B (280-320 nm) and UVA (320-400 nm), induces photocarcinogenesis via various pathways. UV light can directly cause DNA damage, resulting in genetic mutations if not repaired correctly. UV light can also induce photocarcinogenesis by generating reactive oxygen species, inducing immunosuppression and inflammation. Recently, visible light (400-760 nm) has been shown to contribute to photocarcinogenesis by activating oxidative pathways. In addition to the irradiation dose (fluence, J/m2), UVB irradiance (W/m2) is also considered a factor influencing photocarcinogenesis. In this review, we summarize the mechanisms of photocarcinogenesis and provide strategies to prevent skin cancer.
Collapse
Affiliation(s)
- Ting-Ting Yang
- Department of Dermatology, Kaohsiung Medical University Gangshan Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Che E Lan
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
21
|
Fatima M, Al-Keridis LA, Adnan M, Alshammari N, Sulieman AME, Khan MR. Jasminum humile extract mitigates carrageenan-induced paw oedema in rats by modulating inflammatory and antioxidant signalling pathways. Inflammopharmacology 2025; 33:1907-1920. [PMID: 40042724 DOI: 10.1007/s10787-025-01692-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/31/2025] [Indexed: 04/13/2025]
Abstract
BACKGROUND Jasminum humile is widely used in traditional medicines to treat hard lumps, mouth inflammation, ringworms, and other infections. Leaf decoction of the plant is known to be effective in treating various skin conditions. In addition, root juice is traditionally utilized as a remedy for ringworm infections. Studies have reported that J. humile contains various antioxidant metabolites with analgesic and anti-inflammatory properties. In this study, J. humile chloroform extract (JHC) was investigated for anti-inflammatory effects against carrageenan-induced paw oedema in rat models. METHODS High-performance liquid chromatography was used to examine phenolic compounds present in JHC. The in-vivo anti-inflammatory activities were investigated using carrageenan-induced paw oedema rat models, while indomethacin was referred to as positive control. Therapeutic properties of JHC were examined by assessing paw volumes, motility score, and inflammatory proteins in serum. The anti-inflammatory nature of JHC was further investigated by biochemical and hematological profiles along with genetic expression of inflammatory and antioxidant genes through qRT-PCR analysis. RESULTS Indomethacin at 10 mg/kg and JHC at 100, 200, and 300 mg/kg doses decreased the concentration of C-reactive protein (CRP) while upregulating the concentration of albumin and myeloperoxidase (MPO). Moreover, JHC administration reduced the expression levels of inflammatory markers, cyclooxygenase-2 (COX2), and inducible nitric oxide synthase (iNOS) compared to the Carr-treated control. However, a significant rise was induced in nuclear factor erythroid 2-related factor 2 (Nrf-2) and heme oxygenase 1 (HO-1) levels after JHC treatment as compared to Carr-treated rats. CONCLUSION These results showed significant anti-inflammatory potential of J. humile by increasing the activity levels of enzymatic antioxidants and lowering inflammatory markers. These results confirm the beneficial use of natural plants in the development of new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Mehreen Fatima
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, P.O. Box 45320, Islamabad, Pakistan.
| | - Lamya Ahmed Al-Keridis
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | - Nawaf Alshammari
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | | | - Muhammad Rashid Khan
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, P.O. Box 45320, Islamabad, Pakistan
| |
Collapse
|
22
|
Patnaik R, Varghese RL, Banerjee Y. Selective Modulation of PAR-2-Driven Inflammatory Pathways by Oleocanthal: Attenuation of TNF-α and Calcium Dysregulation in Colorectal Cancer Models. Int J Mol Sci 2025; 26:2934. [PMID: 40243559 PMCID: PMC11988659 DOI: 10.3390/ijms26072934] [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: 02/07/2025] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Colorectal cancer (CRC) remains a principal contributor to oncological mortality worldwide, with chronic inflammation serving as a fundamental driver of its pathogenesis. Protease-activated receptor-2 (PAR-2), a G-protein-coupled receptor, orchestrates inflammation-driven tumorigenesis by potentiating NF-κB and Wnt/β-catenin signaling, thereby fostering epithelial-mesenchymal transition (EMT), immune evasion, and therapeutic resistance. Despite its pathological significance, targeted modulation of PAR-2 remains an underexplored avenue in CRC therapeutics. Oleocanthal (OC), a phenolic constituent of extra virgin olive oil, is recognized for its potent anti-inflammatory and anti-cancer properties; however, its regulatory influence on PAR-2 signaling in CRC is yet to be elucidated. This study interrogates the impact of OC on PAR-2-mediated inflammatory cascades using HT-29 and Caco-2 CRC cell lines subjected to lipopolysaccharide (LPS)-induced activation of PAR-2. Expression levels of PAR-2 and TNF-α were quantified through Western blotting and RT-PCR, while ELISA assessed TNF-α secretion. Intracellular calcium flux, a pivotal modulator of PAR-2-driven oncogenic inflammation, was evaluated via Fluo-4 calcium assays. LPS markedly elevated PAR-2 expression at both mRNA and protein levels in CRC cells (p < 0.01, one-way ANOVA). OC administration (20-150 μg/mL) elicited a dose-dependent suppression of PAR-2, with maximal inhibition at 100-150 μg/mL (p < 0.001, Tukey's post hoc test). Concomitant reductions in TNF-α transcription (p < 0.01) and secretion (p < 0.001) were observed, corroborating the anti-inflammatory efficacy of OC. Additionally, OC ameliorated LPS-induced calcium dysregulation, restoring intracellular calcium homeostasis in a concentration-dependent manner (p < 0.01). Crucially, OC exhibited selectivity for PAR-2, leaving PAR-1 expression unaltered (p > 0.05), underscoring its precision as a therapeutic agent. These findings position OC as a selective modulator of PAR-2-driven inflammation in CRC, disrupting the pro-tumorigenic microenvironment through attenuation of TNF-α secretion, calcium dysregulation, and oncogenic signaling pathways. This study furnishes mechanistic insights into OC's potential as a nutraceutical intervention in inflammation-associated CRC. Given the variability in OC bioavailability and content in commercial olive oil, future investigations should delineate optimal dosing strategies and in vivo efficacy to advance its translational potential in CRC therapy.
Collapse
Affiliation(s)
- Rajashree Patnaik
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates; (R.P.); (R.L.V.)
| | - Riah Lee Varghese
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates; (R.P.); (R.L.V.)
| | - Yajnavalka Banerjee
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates; (R.P.); (R.L.V.)
- Centre for Medical Education, School of Medicine, University of Dundee Ninewells Hospital Dundee, Dundee DD2 1SG, UK
| |
Collapse
|
23
|
Liu Z, Zhu H, Zhang F, Huang W, Zhu S, He S, Yao Y, Song Q, Zhang X. Dendritic cell-derived MYD88 potentiates as a biomarker for immune regulation in hepatocellular carcinoma and may predict a better immunological result. Front Cell Dev Biol 2025; 13:1554705. [PMID: 40196847 PMCID: PMC11973264 DOI: 10.3389/fcell.2025.1554705] [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: 01/02/2025] [Accepted: 03/12/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction MYD88 (myeloid differentiation primary response 88) is a key adaptor protein mediate immune responses, primarily through Toll-like receptors (TLRs) and interleukin-1 receptor (IL-1R) signaling. The TLR/MYD88 pathway plays a critical role in dendritic cells (DC) maturation and function, contributing to the body's innate immunity. Recent studies have further highlighted MYD88's pivotal role in intrinsic immunity and its regulatory influence on the tumor microenvironment (TME) in hepatocellular carcinoma (HCC). The expression of MYD88 in DCs and its regulatory role in the TME have gained increasing attention. Methods RNA-sequencing data retrieved from the TCGA and GEO databases were utilized for both the training and validation of our signature. Single-cell RNA transcriptome data from GEO were analyzed to investigate the correlation among subclusters of T cells, myeloid cells, and dendritic cells (DCs) within the HCC tumor microenvironment (TME). A combination of bioinformatics and machine learning approaches was employed to perform statistical analyses.Additionally, flow cytometry was conducted to quantify T cell subtypes and assess biomarker expression in DCs. A BALB/c-derived xenograft mouse model was established to evaluate the functional role of MyD88 in tumor progression and immunotherapy response. Furthermore, immunohistochemical (IHC) staining was performed to reassess the biological effects of MyD88 in HCC patients undergoing immune checkpoint inhibitor (ICI) therapy. Results Our pan-cancer data analysis further highlights the significant impact of MYD88 on clinical outcomes in HCC. Analysis of TCGA and GEO databases confirms that MYD88 serves as a key signaling molecule in DCs, reinforcing its critical role in immune regulation. Our in vitro experiments demonstrates that MyD88 modulates T cell function through DCs. In vivo, H22 tumor cells exhibited accelerated growth in MyD88 knockout mice and a reduced response to anti-PD-1 treatment, whereas wild-type mice showed the opposite trend. Discussion These findings underscore the critical role of MYD88 in DC function, suggesting its potential as a biomarker for immunoregulation in HCC. By shaping the TME, MYD88 not only regulates the immune response in HCC but also influences patient clinical outcomes. Both ex vivo and in vivo experiments further validate that MYD88 impacts DC functionality, contributing to variations in HCC progression.
Collapse
Affiliation(s)
- Zheming Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
- Wuhan University Shenzhen Research Institute, Shenzhen, China
| | - Hengbo Zhu
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Fengxia Zhang
- Department of Rehabilitation Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenting Huang
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shipeng Zhu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Songjiang He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xue Zhang
- Department of Breast, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
24
|
Xu P, Zhang Z, Zhu H, Hu C, Ying Y. Causal role of immune cells in head and neck squamous cell carcinoma. Discov Oncol 2025; 16:372. [PMID: 40119021 PMCID: PMC11928708 DOI: 10.1007/s12672-025-02135-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/12/2025] [Indexed: 03/24/2025] Open
Abstract
Chronic inflammation, immune cell infiltration, and metabolic reprogramming play a crucial role in the development of head and neck squamous cell carcinoma (HNSCC). Based on the summary-level data from a genome-wide association study (GWAS), this study used Mendelian randomization (MR) analysis to investigate the causal relationship between immune cell phenotype and HNSCC. Two-step MR was employed to quantify the proportion of the effect of metabolite-mediated immunophenotypes on HNSCC. 16 immune phenotypes exhibited a causal relationship with HNSCC. HLA DR + CD4 + AC (OR: 1.31, 95% CI 1.07-1.59, P < 0.01) and EM CD8br AC cells (OR: 1.36, 95% CI 1.14-1.62, P < 0.01) showed the most significant P values. 13 metabolites have a causal relationship with HNSCC (P < 0.05). The proportion of the effect of Bilirubin degradation product, C16H18N2O5 (3) levels-mediated HLA DR + CD4 + AC on HNSCC was 7.17% (95% CI 1.04%-13.3%), while the proportion of the effect of Glutamine to alanine ratio-mediated EM CD8br AC on HNSCC was 8.88% (95% CI 2.2%-15.6%). Moreover, single-cell RNA-sequencing analysis confirmed that HLA DR + CD4 + AC and EM CD8br AC cells were commonly present in the HNSCC tumor microenvironment (TME). High expression of two immune phenotypes indicated a poorer prognosis. The results of differentially expressed genes (DEGs) analysis and functional enrichment indicated that Interferon-induced protein with tetratricopeptide repeats 1 (IFIT1)/Follicular dendritic cell secreted protein (FDCSP) and Keratin 19 type I (KRT19)/Kallikrein-related peptidase 5 (KLK5) may be responsible genes for two immune phenotypes, respectively. The results of quantitative real-time polymerase chain reaction (qRT-PCR) and survival analysis showed that FDCSP was significantly downregulated in HNSCC and could exert a protective effect. Collectively, our study clarified the causal relationship between immune cells, metabolites, and HNSCC, providing new insight for clinical diagnosis and treatment of HNSCC.
Collapse
Affiliation(s)
- Panshu Xu
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang, People's Republic of China
| | - Zhenxing Zhang
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang, People's Republic of China
| | - Haoran Zhu
- Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Conglin Hu
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang, People's Republic of China
| | - Yukang Ying
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang, People's Republic of China.
| |
Collapse
|
25
|
Kiran S, Xue Y, Sarker DB, Sang QXA. Effects of Induced Pluripotent Stem Cell-Derived Astrocytes on Cisplatin Sensitivity in Pediatric Brain Cancer Cells. Cancers (Basel) 2025; 17:997. [PMID: 40149331 PMCID: PMC11940393 DOI: 10.3390/cancers17060997] [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: 01/27/2025] [Revised: 03/06/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Background: ATRTs and DIPGs are deadly pediatric brain tumors with poor prognosis. These tumors can develop resistance to chemotherapies, which may be significantly influenced by their microenvironment. Since astrocytes are the most abundant glial cell type in the brain microenvironment and may support tumor growth and chemoresistance, this study investigated the effects of induced pluripotent stem cell-derived astrocytes (iPSC-astrocytes) on cisplatin sensitivity in CHLA-05-ATRT and SF8628 (DIPG) cells. iPSCs provide an unlimited and standardized source of nascent astrocytes, which enables modeling the interaction between childhood brain tumor cells and iPSC-astrocytes within a controlled coculture system. Methods: To study the effects on tumor growth, the iPSC-astrocytes were cocultured with tumor cells. Additionally, the tumor cells were exposed to various concentrations of cisplatin to evaluate their chemosensitivity in the presence of astrocytes. Results: The paracrine interaction of iPSC-astrocytes with tumor cells upregulated astrocyte activation markers GFAP and STAT3 and promoted tumor cell proliferation. Moreover, the cisplatin treatment significantly decreased the viability of CHLA-05-ATRT and SF8628 cells. However, tumor cells exhibited reduced sensitivity to cisplatin in the coculture with iPSC-astrocytes. During cisplatin treatment, DIPG cells in particular showed upregulation of resistance markers, ERK1, STAT3, and MTDH, which are associated with enhanced proliferation and invasion. They also had increased expression of APEX1, which is involved in the base excision repair pathway following cisplatin-induced DNA damage. Conclusion: These findings underscore the significance of the tumor microenvironment in modulating tumor cell survival and chemosensitivity.
Collapse
Affiliation(s)
- Sonia Kiran
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Yu Xue
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Drishty B. Sarker
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
26
|
You F, Zhang H, Meng L, Li C, Yang Y, Wang Y, Zhao R, Chao L. Mechanistic investigation of Shuanghuanglian against infectious bronchitis in chickens: a network pharmacology and molecular dynamics study. Front Vet Sci 2025; 12:1557850. [PMID: 40144526 PMCID: PMC11936991 DOI: 10.3389/fvets.2025.1557850] [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: 01/09/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction Infectious bronchitis (IB) poses a major challenge to global poultry production, causing substantial economic burdens and underscoring the necessity for novel therapeutic interventions given the limitations of current vaccines and conventional antiviral agents. The purpose of this study is to comprehensively explore the active components in Shuanghuanglian and their interaction with the key pathological targets of IBV (Infectious bronchitis virus) infection. By using advanced computational methods, this study aims not only to identify the therapeutic potential of active ingredients, but also to reveal their mechanism of action against IBV. Methods Through integrative systems pharmacology approaches, we systematically investigated Shuanghuanglian and its phytochemical constituents against IB, employing multi-omics analysis, ensemble machine learning, and all-atom molecular dynamics (MD) simulations. Network pharmacology revealed 65 target genes associated with Shuanghuanglian's primary bioactive components (quercetin, kaempferol, wogonin, and luteolin), exhibiting high network centrality. Results Using the TCMSP database, we found 65 target genes associated with key active components, such as quercetin and kaempferol, which exhibited strong connectivity in our network analysis. The GeneCards database also identified 40 common target genes shared by Shuanghuanglian and IB. Importantly, BCL2 and IL6 were recognized as key targets in the protein-protein interaction (PPI) network analysis, highlighting their roles in apoptosis and inflammation. Furthermore, analyses using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways revealed significant roles in regulating the cell cycle and inflammatory responses. Machine learning techniques identified BCL2 and IL6 as critical genes for therapeutic intervention, supported by molecular docking results that showed strong binding energies. Furthermore, molecular dynamics simulations confirm the stability of the complexes, underscoring the importance of these interactions for treatment efficacy. Conclusion We used a variety of analytical methods, and finally identified the potential active ingredients of Shuanghuanglian as kaempferol, quercetin, wogonin, and luteolin. The active ingredients target BCL2 and IL6 and play a therapeutic role in avian infectious bronchitis by inhibiting apoptosis and reducing inflammatory response.
Collapse
Affiliation(s)
- Fuming You
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Hanzhao Zhang
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Linghao Meng
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Chuanhong Li
- College of Computer Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Yuxia Yang
- College of Computer Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | | | - Rigetu Zhao
- Chifeng Academy of Agricultural and Animal Husbandry Sciences, Chifeng, China
| | - Luomeng Chao
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
- Inner Mongolia Rambo Testing Technology Limited Company, Tongliao, China
- Inner Mongolia Engineering Technology Research Center for Prevention and Control of Beef Cattle Diseases, Tongliao, China
| |
Collapse
|
27
|
Dragomir RD, Negru AG, Mercioni MA, Popovici D, Săftescu S, Blidari AR, Curcă RO, Sas I. The Role of Initial Neutropenia and Neutrophil Dynamics in Personalizing Chemotherapy for Platinum-Resistant Ovarian Cancer. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:470. [PMID: 40142281 PMCID: PMC11944004 DOI: 10.3390/medicina61030470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025]
Abstract
Background and Objectives: Platinum-resistant ovarian cancer (PROC) is associated with limited treatment options and poor outcomes, with median progression-free survival (PFS) and overall survival (OS) remaining suboptimal. Neutropenia, a common chemotherapy-related toxicity, has shown potential as a predictive biomarker for treatment efficacy in several malignancies, including ovarian cancer. However, its role as a prognostic marker, particularly baseline neutropenia, remains underexplored. This study aimed to evaluate the prognostic and predictive value of initial neutropenia and neutrophil dynamics in PROC patients undergoing chemotherapy. Materials and Methods: A retrospective cohort study was conducted on 250 PROC patients treated between 2018 and 2022 at the OncoHelp Medical Center, Timișoara, Romania. Patients were stratified into two groups based on baseline absolute neutrophil count (ANC), as those with initial neutropenia (ANC < 2000/mm3) and without initial neutropenia (ANC ≥ 2000/mm3). Clinical outcomes, including tumor response, PFS, and OS, were assessed using RECIST 1.1 criteria. Hematological toxicities and neutrophil dynamics across three chemotherapy cycles were analyzed. Results: Patients with baseline neutropenia demonstrated significantly higher tumor response rates (47.05% vs. 27.27%; p = 0.002), longer median PFS (8.2 vs. 6.3 months; p = 0.008), and extended median OS (14.5 vs. 11.2 months; p = 0.002). Hematological toxicities, including Grade ≥3 neutropenia and febrile neutropenia, were more frequent in the neutropenic group (p < 0.001). Baseline ANC thresholds effectively predicted clinical outcomes, with an AUC of 0.79 for OS. Conclusions: Baseline neutropenia is a significant prognostic marker in PROC, correlating with improved tumor response and survival outcomes despite increased hematological toxicities. These findings support incorporating baseline ANC into treatment personalization strategies for PROC.
Collapse
Affiliation(s)
- Radu-Dumitru Dragomir
- Department of Obstetrics and Gynecology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (R.-D.D.); (I.S.)
| | - Alina-Gabriela Negru
- Department of Cardiology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Marina-Adriana Mercioni
- Faculty of Electronics, Telecommunications and Information Technologies, Politehnica University Timisoara, 300223 Timișoara, Romania;
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Dorel Popovici
- Department of Oncology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (D.P.); (S.S.)
| | - Sorin Săftescu
- Department of Oncology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (D.P.); (S.S.)
| | - Andiana Roxana Blidari
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | | | - Ioan Sas
- Department of Obstetrics and Gynecology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (R.-D.D.); (I.S.)
| |
Collapse
|
28
|
Yang J, Zhou F, Luo X, Fang Y, Wang X, Liu X, Xiao R, Jiang D, Tang Y, Yang G, You L, Zhao Y. Enhancer reprogramming: critical roles in cancer and promising therapeutic strategies. Cell Death Discov 2025; 11:84. [PMID: 40032852 DOI: 10.1038/s41420-025-02366-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 01/24/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025] Open
Abstract
Transcriptional dysregulation is a hallmark of cancer initiation and progression, driven by genetic and epigenetic alterations. Enhancer reprogramming has emerged as a pivotal driver of carcinogenesis, with cancer cells often relying on aberrant transcriptional programs. The advent of high-throughput sequencing technologies has provided critical insights into enhancer reprogramming events and their role in malignancy. While targeting enhancers presents a promising therapeutic strategy, significant challenges remain. These include the off-target effects of enhancer-targeting technologies, the complexity and redundancy of enhancer networks, and the dynamic nature of enhancer reprogramming, which may contribute to therapeutic resistance. This review comprehensively encapsulates the structural attributes of enhancers, delineates the mechanisms underlying their dysregulation in malignant transformation, and evaluates the therapeutic opportunities and limitations associated with targeting enhancers in cancer.
Collapse
Affiliation(s)
- Jinshou Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Feihan Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xiyuan Luo
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Yuan Fang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Ruiling Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Decheng Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Yuemeng Tang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| |
Collapse
|
29
|
Xiang L, Shen Y, Liu S, Fan B, Zhan J, Zhou Y, Jiang B, Wang M, Liu Q, Liu X, Zou Y, Sun S. Guggulsterone ameliorates psoriasis by inhibiting keratinocyte proliferation and inflammation through induction of miR-17 directly targeting JAK1 and STAT3. Biochem Pharmacol 2025; 233:116745. [PMID: 39793717 DOI: 10.1016/j.bcp.2025.116745] [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: 08/17/2024] [Revised: 11/30/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
The pathogenesis of psoriasis involves hyperproliferation of epidermal keratinocytes and abnormal interactions between activated keratinocytes and infiltrating immune cells. Emerging evidence has shown that keratinocytes play essential roles in both the initiation and maintenance of psoriasis, suggesting that exposing keratinocytes to agents with antiproliferative and anti-inflammatory effects may be effective for psoriasis treatment. Guggulsterone (GS), a plant sterol derived from the gum resin of Commiphora wightii, possesses a variety of pharmacological activities. However, the effects of GS on psoriasis and the underlying mechanism have not been elucidated. In this study, we evaluated the therapeutic effect of GS on psoriasis using an imiquimod-induced psoriasis mouse model and investigated the effect of GS on human keratinocytes and the underlying mechanism. We found that GS effectively alleviated psoriasis-like skin lesions in imiquimod-induced psoriasis model mice and that GS suppressed the proliferation, migration, and production of proinflammatory cytokines, chemokines and antimicrobial peptides in keratinocytes. Transcriptome analysis by RNA-seq revealed that the differentially expressed genes (DEGs) induced by GS in keratinocytes were intricately linked to the pathogenesis of psoriasis. Furthermore, STAT3, a key player in the development and pathogenesis of psoriasis, was identified as a critical downstream mediator of GS in keratinocytes. Mechanistically, GS upregulated the expression of miR-17-5p, which directly binds to the 3'-untranslated regions (3'UTRs) of JAK1 and STAT3, leading to the downregulation of JAK1 and STAT3 expression. Collectively, these findings suggest that GS may serve as an effective natural compound for the treatment of psoriasis.
Collapse
Affiliation(s)
- Lu Xiang
- Department of Dermatology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan 250011, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yangli Shen
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Shuangteng Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Bowen Fan
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Jiafeng Zhan
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yadi Zhou
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Molin Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Qiao Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Xiaofei Liu
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China.
| | - Shuna Sun
- Department of Dermatology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan 250011, China.
| |
Collapse
|
30
|
Luo M, Jin T, Fang Y, Chen F, Zhu L, Bai J, Ding J. Signaling Pathways Involved in Acute Pancreatitis. J Inflamm Res 2025; 18:2287-2303. [PMID: 40230438 PMCID: PMC11995411 DOI: 10.2147/jir.s485804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 01/25/2025] [Indexed: 04/16/2025] Open
Abstract
Acute pancreatitis (AP) is a common digestive emergency with high morbidity and mortality. Over the past decade, significant progress has been made in understanding the mechanisms of AP, including oxidative stress, disruptions in calcium homeostasis, endoplasmic reticulum stress, inflammatory responses, and various forms of cell death. This review provides an overview of the typical signaling pathways involved and proposes the latest clinical translation prospects. These strategies are important for the early management of AP, preventing multi-organ injury, and improving the overall prognosis of the disease.
Collapse
Affiliation(s)
- Mengchen Luo
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Ting Jin
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Yi Fang
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Feng Chen
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Lujian Zhu
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Jin Ding
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| |
Collapse
|
31
|
Zhou S, Wen H, He X, Han X, Li H. Pulsed electromagnetic field ameliorates the progression of osteoarthritis via the Sirt1/NF-κB pathway. Arthritis Res Ther 2025; 27:33. [PMID: 39953605 PMCID: PMC11827477 DOI: 10.1186/s13075-025-03492-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/30/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Pulsed electromagnetic field (PEMF) is a non-invasive treatment that utilizes electromagnetic fields to reduce inflammation and promote tissue repair. However, PEMFs' anti-inflammatory effect on osteoarthritis (OA) and the potential mechanism has not been fully elucidated. METHODS Human chondrocytes (C28/I2) were stimulated with interleukin (IL)-1β with or without the treatment of PEMF. CCK-8 assay Kit was used to detect cell viability. RT-qPCR, ELISA, immunofluorescent staining and western blot was used to analyze relative markers of inflammatory response and extracellular matrix (ECM) under the treatment of PEMF and related mechanism. Besides, the significance role of Sirt1 was assessed by using the Sirt1 inhibitor (EX-527). Moreover, immunohistochemistry and immunofluorescence staining were carried out to evaluate the curative effect of PEMF on OA mice induced by the destabilization of the medial meniscus (DMM). RESULTS PEMF inhibited IL-1β-mediated the expression of pro-inflammatory factors. Besides, PEMF alleviated IL-1β-induced degradation of ECM by increasing the expression of Col2a1 and ACAN, while inhibiting the expression of MMP13 and ADAMTS5. At the mechanism level, PEMF increased the expression of Sirt1 and inhibited IL-1β-induced the activation of NF-κB pathway. Furthermore, blocking Sirt1 with EX-527 attenuated the effect of PEMF on the inhibition of NF-κB pathway and the expression of ECM in IL-1β-induced chondrocytes. In vivo, PEMF-treated OA mice showed low modified mankin scores, reduced the number of osteophytes and preserved joint structure. CONCLUSIONS Our results suggest that PEMF inhibits NF-κB pathway and blocks the expression of inflammatory factors by activating the expression of Sirt1, which may be a novel strategy for OA.
Collapse
Affiliation(s)
- Siqi Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Haiyan Wen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiongwei He
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaotao Han
- Wuhan National High Magnetic Field Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- State Key Laboratory of Advanced Electromagnetic Engineering and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Haohuan Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
32
|
Worley J, Noh H, You D, Turunen MM, Ding H, Paull E, Griffin AT, Grunn A, Zhang M, Guillan K, Bush EC, Brosius SJ, Hibshoosh H, Mundi PS, Sims P, Dalerba P, Dela Cruz FS, Kung AL, Califano A. Identification and Pharmacological Targeting of Treatment-Resistant, Stem-like Breast Cancer Cells for Combination Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.08.562798. [PMID: 38798673 PMCID: PMC11118419 DOI: 10.1101/2023.11.08.562798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Tumors frequently harbor isogenic yet epigenetically distinct subpopulations of multi-potent cells with high tumor-initiating potential-often called Cancer Stem-Like Cells (CSLCs). These can display preferential resistance to standard-of-care chemotherapy. Single-cell analyses can help elucidate Master Regulator (MR) proteins responsible for governing the transcriptional state of these cells, thus revealing complementary dependencies that may be leveraged via combination therapy. Interrogation of single-cell RNA sequencing profiles from seven metastatic breast cancer patients, using perturbational profiles of clinically relevant drugs, identified drugs predicted to invert the activity of MR proteins governing the transcriptional state of chemoresistant CSLCs, which were then validated by CROP-seq assays. The top drug, the anthelmintic albendazole, depleted this subpopulation in vivo without noticeable cytotoxicity. Moreover, sequential cycles of albendazole and paclitaxel-a commonly used chemotherapeutic -displayed significant synergy in a patient-derived xenograft (PDX) from a TNBC patient, suggesting that network-based approaches can help develop mechanism-based combinatorial therapies targeting complementary subpopulations. Statement of significance Network-based approaches, as shown in a study on metastatic breast cancer, can develop effective combinatorial therapies targeting complementary subpopulations. By analyzing scRNA-seq data and using clinically relevant drugs, researchers identified and depleted chemoresistant Cancer Stem-Like Cells, enhancing the efficacy of standard chemotherapies.
Collapse
Affiliation(s)
- Jeremy Worley
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- J.P. Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Heeju Noh
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Daoqi You
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mikko M Turunen
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Hongxu Ding
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- Department of Pharmacy Practice & Science, College of Pharmacy, University of Arizona, Tucson, Arizona, USA 85721
| | - Evan Paull
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Aaron T Griffin
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Adina Grunn
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Mingxuan Zhang
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Kristina Guillan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Erin C Bush
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Samantha J Brosius
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA 10032
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, USA 10032
| | - Prabhjot S Mundi
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA 10032
| | - Peter Sims
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Piero Dalerba
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA 10032
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, USA 10032
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, USA 10032
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
| | - Filemon S Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrew L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea Califano
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA 10032
- Department of Biochemistry & Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA 10032
- J.P. Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY USA 10032
| |
Collapse
|
33
|
Wang Y, Li D, Zhao L, Liu J, Dou D, Liu N, Zhuo Y, Zhang S. Mechanism of Yinxu Weitong Capsule in the treatment of precancerous lesions of gastric cancer based on network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119303. [PMID: 39761837 DOI: 10.1016/j.jep.2024.119303] [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: 10/19/2024] [Revised: 12/13/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yinxu Weitong Capsule (YXWTC) is a Chinese patent medicine used to treat chronic gastritis. However, its efficacy and mechanisms of action in treating precancerous lesions of gastric cancer (PLGC) remain unclear. AIM OF THE STUDY To evaluate the effects of YXWTC on PLGC and explore the underlying mechanisms. MATERIALS AND METHODS YXWTC components were identified using ultra-high-performance liquid chromatography coupled with electrospray ionization quadrupole-exactive orbitrap mass spectrometry. A PLGC animal model was established and the protective effects of YXWTC on the gastric mucosa in PLGC rats were evaluated using hematoxylin and eosin (H&E), Alcian blue-periodic acid-Schiff and Alcian blue-high iron diamine staining, and transmission electron microscopy (TEM). The vital organs of the rats were examined using H&E staining to evaluate biosafety. Network pharmacology identified potential targets and pathways of YXWTC in PLGC treatment, followed by molecular docking validation. Various techniques, including enzyme-linked immunosorbent assay, real-time quantitative reverse transcription PCR, Western blotting, immunohistochemistry, apoptosis detection, and reactive oxygen species fluorescence staining were employed to elucidate the underlying mechanisms. RESULTS In total, 340 YXWTC components were identified. YXWTC effectively improves gastric mucosal pathology in rats with PLGC. Network pharmacology identified 403 targets common to PLGC and YXWTC. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses identified 2,323 biological processes and 206 signaling pathways, respectively. Molecular docking revealed that the primary target proteins and major drug molecules exhibited strong binding affinities. Animal studies demonstrated that YXWTC inhibited the IL-6/STAT3 pathway, promoted mitochondrial apoptosis, and induced ROS release. CONCLUSIONS We verified the pharmacodynamic effects of YXWTC in PLGC. In summary, the effects are mediated by inhibition of the IL-6/STAT3 pathway, promotion of mitochondrial apoptosis, and induction of ROS release.
Collapse
Affiliation(s)
- Yichong Wang
- Digestive Disease Center, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23, Back Street of Art Museum, Dongcheng District, Beijing, 100010, China.
| | - Danyan Li
- Digestive Disease Center, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23, Back Street of Art Museum, Dongcheng District, Beijing, 100010, China.
| | - Luqing Zhao
- Digestive Disease Center, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23, Back Street of Art Museum, Dongcheng District, Beijing, 100010, China.
| | - Jixiang Liu
- Digestive Disease Center, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23, Back Street of Art Museum, Dongcheng District, Beijing, 100010, China; Beijing University of Chinese Medicine, 11 North Third Ring East Road, Chaoyang District, Beijing, 100010, China.
| | - Dan Dou
- Digestive Disease Center, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23, Back Street of Art Museum, Dongcheng District, Beijing, 100010, China.
| | - Nian Liu
- Digestive Disease Center, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23, Back Street of Art Museum, Dongcheng District, Beijing, 100010, China; Beijing University of Chinese Medicine, 11 North Third Ring East Road, Chaoyang District, Beijing, 100010, China.
| | - Yudi Zhuo
- Digestive Disease Center, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23, Back Street of Art Museum, Dongcheng District, Beijing, 100010, China; Beijing University of Chinese Medicine, 11 North Third Ring East Road, Chaoyang District, Beijing, 100010, China.
| | - Shengsheng Zhang
- Digestive Disease Center, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23, Back Street of Art Museum, Dongcheng District, Beijing, 100010, China.
| |
Collapse
|
34
|
Qin H, Wang J, Bai L, Ding H, Ding H, Zhang F, Han Y. Aerosol inhalation of rhIL-10 improves acute lung injury in mice by affecting pulmonary neutrophil phenotypes through neutrophil-platelet aggregates. Int Immunopharmacol 2025; 147:113948. [PMID: 39778276 DOI: 10.1016/j.intimp.2024.113948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 01/11/2025]
Abstract
This study investigates the therapeutic effects of recombinant human IL-10 (rhIL-10) administered via aerosol inhalation in acute lung injury (ALI), with a particular focus on neutrophils. It explores how rhIL-10, in the presence of platelets, modulates neutrophil polarization to ameliorate acute lung injury. Initially, the ALI model established in mice demonstrated that aerosol inhalation of rhIL-10 significantly mitigated the cytokine storm in the lungs, reduced pulmonary edema, and alleviated histopathological damage to lung tissue. Additionally, rhIL-10 administration was found to decrease neutrophil infiltration and platelet activation in the lungs of mice, inhibiting the formation of platelet-neutrophil aggregates (PNAs) and promoting the differentiation of neutrophils toward an anti-inflammatory phenotype in the presence of platelets. Subsequently, primary neutrophils and platelets were isolated from mouse bone marrow and blood to explore the underlying mechanisms. The results indicated that rhIL-10 promotes the expression of the signal transducer and activator of transcription 3 (STAT3) and the suppressor of cytokine signaling 3 (SOCS3) in neutrophils while inhibiting the activation of the nuclear factor kappa B (NF-κB) and the NF-κB inhibitor (IκB), which in turn enhances CD40 expression. This interaction facilitates the formation of PNAs and influences neutrophil phenotype differentiation. Furthermore, the application of the STAT3 phosphorylation inhibitor Stattic and CD40 antibody in vivo provided further validation of this potential mechanism. In conclusion, these results indicate that aerosol inhalation of rhIL-10 effectively ameliorates ALI. The underlying mechanism may involve the modulation of the neutrophil STAT/SOCS-IκB/NF-κB-CD40 signaling pathway, promoting interactions between neutrophils and platelets that facilitate the differentiation of neutrophils toward an anti-inflammatory phenotype.
Collapse
Affiliation(s)
- Huan Qin
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jiangang Wang
- School of Basic Medicine, Qingdao University, Qingdao, China; Kanglitai Biopharmaceutical (Qingdao) Co. Ltd., Qingdao, China
| | - Luyuan Bai
- Xianyang Hospital of Yan'an University, Xianyang, China
| | - Huiqin Ding
- School of Basic Medicine, Qingdao University, Qingdao, China; Kanglitai Biopharmaceutical (Qingdao) Co. Ltd., Qingdao, China
| | | | | | - Yantao Han
- School of Basic Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
35
|
Cioanca O, Lungu II, Batir-Marin D, Lungu A, Marin GA, Huzum R, Stefanache A, Sekeroglu N, Hancianu M. Modulating Polyphenol Activity with Metal Ions: Insights into Dermatological Applications. Pharmaceutics 2025; 17:194. [PMID: 40006561 PMCID: PMC11858937 DOI: 10.3390/pharmaceutics17020194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/29/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND The skin represents the first barrier of defense, and its integrity is crucial for overall health. Skin wounds present a considerable risk seeing how their progression is rapid and sometimes they are caused by comorbidities like diabetes and venous diseases. Nutraceutical combinations like the ones between polyphenols and metal ions present considerable applications thanks to their increased bioavailability and their ability to modulate intrinsic molecular pathways. METHODS The research findings presented in this paper are based on a systematic review of the current literature with an emphasis on nanotechnology and regenerative medicine strategies that incorporate polyphenols and metallic nanoparticles (NPs). The key studies which described the action mechanisms, efficacy, and safety of these hybrid formulations were reviewed. RESULTS Nanocomposites of polyphenol and metal promote healing by activating signaling pathways such as PI3K/Akt and ERK1/2, which in turn improve fibroblast migration and proliferation. Nanoparticles of silver and copper have antibacterial, angiogenesis-promoting, inflammation-modulating capabilities. With their ability to induce apoptosis and restrict cell growth, these composites have the potential to cure skin malignancies in addition to facilitating wound healing. CONCLUSIONS Nanocomposites of polyphenols and metals provide hope for the treatment of cancer and chronic wounds. Their antimicrobial capabilities, capacity to modulate inflammatory responses, and enhancement of fibroblast activity all point to their medicinal potential. Furthermore, these composites have the ability to decrease inflammation associated with tumors while simultaneously inducing cell death in cancer cells. Clarifying their mechanisms, guaranteeing stability, and enhancing effective delivery techniques for clinical usage should be the focus of future studies.
Collapse
Affiliation(s)
- Oana Cioanca
- Department of Pharmacognosy, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ionut-Iulian Lungu
- Department of General and Inorganic Chemistry, Faculty of Pharmacy, “Grigore. T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Denisa Batir-Marin
- Department of Pharmaceutical Sciences, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania
| | - Andreea Lungu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 400347 Iasi, Romania
| | - George-Alexandru Marin
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Riana Huzum
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 400347 Iasi, Romania
| | - Alina Stefanache
- Department of General and Inorganic Chemistry, Faculty of Pharmacy, “Grigore. T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Nazim Sekeroglu
- Department of Food Engineering, Faculty of Engineering and Architecture, Kilis 7 Aralık University, 79000 Kilis, Turkey
| | - Monica Hancianu
- Department of Pharmacognosy, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
36
|
García-García VA, Alameda JP, Fernández-Aceñero MJ, Navarro M, García-Escudero R, Page A, Mateo-Gallego R, Paramio JM, Ramírez Á, García-Fernández RA, Bravo A, Casanova ML. Nuclear versus cytoplasmic IKKα signaling in keratinocytes leads to opposite skin phenotypes and inflammatory responses, and a different predisposition to cancer. Oncogene 2025; 44:165-178. [PMID: 39511409 PMCID: PMC11725495 DOI: 10.1038/s41388-024-03203-0] [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: 11/20/2023] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
IKKα is known as an essential protein for skin homeostasis. However, the lack of suitable models to investigate its functions in the skin has led to IKKα being mistakenly considered as a suppressor of non-melanoma skin cancer (NMSC) development. In this study, using our previously generated transgenic mouse models expressing exogenous IKKα in the cytoplasm (C-IKKα mice) or in the nucleus (N-IKKα mice) of basal keratinocytes, we demonstrate that at each subcellular localization, IKKα differently regulates signaling pathways important for maintaining the balance between keratinocyte proliferation and differentiation, and for the cutaneous inflammatory response. In addition, each type of IKKα-transgenic mice shows different predisposition to the development of spontaneous NMSC. Specifically, N-IKKα mice display an atrophic epidermis with exacerbated terminal differentiation, signs of premature skin aging, premalignant lesions, and develop squamous cell carcinomas (SCCs). Conversely, C-IKKα mice, whose keratinocytes are nearly devoid of endogenous nuclear IKKα, do not develop skin SCCs, although they exhibit hyperplastic skin with deficiencies in terminal epidermal differentiation, chronic cutaneous inflammation, and constitutive activation of STAT-3 and NF-κB signaling pathways. Altogether, our data demonstrate that alterations in the localization of IKKα in the nucleus or cytoplasm of keratinocytes cause opposite skin changes and differentially predispose to the growth of skin SCCs.
Collapse
Affiliation(s)
- Verónica A García-García
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Avda. Complutense 40, 28040, Madrid, Spain
- Biomedical Research Institute, University Hospital "12 de Octubre", 28041, Madrid, Spain
| | - Josefa P Alameda
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Avda. Complutense 40, 28040, Madrid, Spain
- Biomedical Research Institute, University Hospital "12 de Octubre", 28041, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | | | - Manuel Navarro
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Avda. Complutense 40, 28040, Madrid, Spain
- Biomedical Research Institute, University Hospital "12 de Octubre", 28041, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Ramón García-Escudero
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Avda. Complutense 40, 28040, Madrid, Spain
- Biomedical Research Institute, University Hospital "12 de Octubre", 28041, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Angustias Page
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Avda. Complutense 40, 28040, Madrid, Spain
- Biomedical Research Institute, University Hospital "12 de Octubre", 28041, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Raúl Mateo-Gallego
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Avda. Complutense 40, 28040, Madrid, Spain
- Biomedical Research Institute, University Hospital "12 de Octubre", 28041, Madrid, Spain
| | - Jesús M Paramio
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Avda. Complutense 40, 28040, Madrid, Spain
- Biomedical Research Institute, University Hospital "12 de Octubre", 28041, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Ángel Ramírez
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Avda. Complutense 40, 28040, Madrid, Spain
- Biomedical Research Institute, University Hospital "12 de Octubre", 28041, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Rosa A García-Fernández
- Department of Animal Medicine and Surgery, Facultad de Veterinaria, UCM, 28040, Madrid, Spain
| | - Ana Bravo
- Department of Anatomy, Animal Production and Veterinary Clinical Sciences, Faculty of Veterinary Medicine, University of Santiago de Compostela, Lugo, Spain
| | - M Llanos Casanova
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Avda. Complutense 40, 28040, Madrid, Spain.
- Biomedical Research Institute, University Hospital "12 de Octubre", 28041, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain.
| |
Collapse
|
37
|
de Freitas S, Makiyama EN, Neves BRO, Pizzolato Cezar ISR, Gonçalves CEDS, Rogero MM, Fock RA. The Influence of Cyanidin-3-Glucoside on the Modulation of Immune Cell Responses by Mesenchymal Stem Cell-Conditioned Medium. Cell Biochem Funct 2025; 43:e70059. [PMID: 39963824 DOI: 10.1002/cbf.70059] [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: 10/14/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 05/10/2025]
Abstract
Mesenchymal stem cells (MSCs) are emerging as promising therapeutic agents due to their immunomodulatory effects, primarily mediated via paracrine signaling. Similarly, anthocyanins, such as cyanidin-3-glucoside (C3G), have demonstrated significant anti-inflammatory properties. In this context, this study investigated the immunomodulatory potential of C3G on MSCs, and subsequent effects on macrophage and lymphocyte responses. Cytotoxicity assays identified 50 µM as the highest nontoxic C3G concentration for MSCs. Flow cytometry confirmed that C3G treatment did not affect MSC viability or cell cycle distribution, even under LPS stimulation. Cytokine production by MSCs was evaluated after treatment with C3G and LPS. While no significant changes were observed in IL-6, IL-10, TGF-β, or PGE2 levels, IL-1β production was significantly reduced in LPS-stimulated MSCs treated with C3G. Protein expression analysis revealed decreased NFκB phosphorylation in LPS-stimulated MSCs treated with C3G, with no changes detected in STAT-3 or PCNA expression. The immunomodulatory effects of MSC-derived conditioned media on macrophages and lymphocytes were also assessed. In LPS-stimulated macrophages, conditioned media from MSCs reduced the production of IL-1β, IL-6, and IL-12. Interestingly, conditioned media from C3G-treated MSCs specifically decreased TNF-α levels, enhanced IL-10 secretion, and further inhibited NFκB phosphorylation. In LPS-stimulated lymphocytes, conditioned media from C3G-treated MSCs suppressed IL-2 production while increasing IL-10 levels. In summary, these findings demonstrate that conditioned media from C3G-treated MSCs modulates immune cell responses more effectively than C3G alone. C3G influences the paracrine activity of MSCs, resulting in a shift in the secretory profile and subsequent effects on immune cell behavior.
Collapse
Affiliation(s)
- Sumara de Freitas
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edson Naoto Makiyama
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Bruna Roberta Oliveira Neves
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Ricardo Ambrósio Fock
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Wang W, He L, Lin T, Xiang F, Wu Y, Zhou F, He Y. Homoharringtonine: mechanisms, clinical applications and research progress. Front Oncol 2025; 15:1522273. [PMID: 39949739 PMCID: PMC11821653 DOI: 10.3389/fonc.2025.1522273] [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: 11/04/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Abstract
Homoharringtonine is a natural alkaloid with significant pharmacological potential that has demonstrated promising efficacy in the treatment of hematological malignancies in recent years. This article systematically reviews the pharmacological mechanisms of Homoharringtonine, focusing on its key roles in inducing apoptosis, inhibiting cell cycle progression, and reducing cell migration and invasion. Additionally, HHT exhibits multiple biological activities, including immunomodulation, antiviral effects, and anti-fibrotic properties, with recent studies also revealing its potential neuroprotective functions. In clinical trials, Homoharringtonine has demonstrated promising efficacy in the treatment of hematological malignancies, particularly in various types such as acute myeloid leukemia and chronic myeloid leukemia. Despite the significant antitumor effects observed in clinical applications, its low bioavailability and potential side effects remain major challenges that limit its widespread use. This article details the latest research advancements aimed at enhancing the bioavailability of Homoharringtonine, including various drug delivery systems such as nanoparticles and liposomes, as well as chemical modification strategies. These approaches not only improve HHT's bioavailability in vivo but also enhance its targeting ability while reducing toxicity to normal cells. Furthermore, the combination of HHT with other drugs presents broader prospects for clinical treatment. By exploring the diverse pharmacological activities of Homoharringtonine in depth, this article aims to provide a foundation for developing novel therapeutic approaches based on natural products, thereby advancing HHT's application research in cancer treatment and other fields.
Collapse
Affiliation(s)
- Wen Wang
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Lan He
- Hunan Provincial Engineering and Technological Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Chinese Medicine and Protecting Visual Function, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Ting Lin
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Engineering and Technological Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Chinese Medicine and Protecting Visual Function, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Fulan Xiang
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Yibin Wu
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Fangliang Zhou
- Hunan Provincial Engineering and Technological Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Chinese Medicine and Protecting Visual Function, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Yingchun He
- Hunan Provincial Engineering and Technological Research Center for Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Chinese Medicine and Protecting Visual Function, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
39
|
Sheng J, You X, Nie D, Fu Y, Ling Q, Yang X, Chen Y, Ma L, Hu S. ZDHHC2 promoted antimycobacterial responses by selective autophagic degradation of B-RAF and C-RAF in macrophages. SCIENCE ADVANCES 2025; 11:eadq7706. [PMID: 39854453 PMCID: PMC11758995 DOI: 10.1126/sciadv.adq7706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025]
Abstract
S-Palmitoylation is a reversible post-translational modification involving saturated fatty acid palmitate-to-cysteine linkage in the protein, which guides many aspects of macrophage physiology in health and disease. However, the precise role and underlying mechanisms of palmitoylation in Mycobacterium tuberculosis infection of macrophages remain elusive. Here, we found that M. tuberculosis infection induced the expression of zinc-finger DHHC domain-type palmitoyl-transferases (ZDHHCs), particularly ZDHHC2, in mouse macrophages. Furthermore, ZDHHC2 deficiency in mouse macrophages impaired the immunity against M. tuberculosis and reduced the production of various proinflammatory cytokines. Mechanistic studies revealed the involvement of ZDHHC2 in mediating the palmitoylation of B-RAF and C-RAF, affecting their autophagic degradation and stabilizing protein levels. The increased abundance of B-RAF and C-RAF subsequently increases the activity of the extracellular signal-regulated kinase (ERK) signaling pathway, affecting the survival of M. tuberculosis within macrophages. These findings suggest that ZDHHC2 is a potential target for treating tuberculosis.
Collapse
Affiliation(s)
- Junli Sheng
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510515, China
| | - Xiaolong You
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510515, China
| | - Dingnai Nie
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510515, China
| | - Yuling Fu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510515, China
| | - Qiao Ling
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510515, China
| | - Xiaodan Yang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510515, China
| | - Yitian Chen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510515, China
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
- Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, China
| | - Shengfeng Hu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510515, China
| |
Collapse
|
40
|
Filipovich E, Gorodkova E, Shcherbakova A, Asaad W, Popov S, Melnichenko G, Mokrysheva N, Utkina M. The role of cell cycle-related genes in the tumorigenesis of adrenal and thyroid neuroendocrine tumors. Heliyon 2025; 11:e41457. [PMID: 39834406 PMCID: PMC11742855 DOI: 10.1016/j.heliyon.2024.e41457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/22/2025] Open
Abstract
The molecular mechanisms underlying adrenal and thyroid neuroendocrine tumors, including their tumorigenesis, progression, and metastasis, involve unique pathways regulating cell cycle progression. To better understand these mechanisms and pathways, extensive in-depth research on cell cycle-related genes is necessary. This review aims to describe and interpret current single-cell RNA sequencing studies on neuroblastoma, medullary thyroid cancer, and pheochromocytoma tumors. Our review summarizes differentially expressed cell cycle-related genes with distinct functions, highlighting their potential as therapeutic targets and components of panels used to determine tumor type or aggressiveness. Although some insights have been gained, there is still limited information on these topics, and further research is required to explore the regulatory mechanisms of these tumors.
Collapse
Affiliation(s)
- Ekaterina Filipovich
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Ekaterina Gorodkova
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Anastasia Shcherbakova
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Walaa Asaad
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Sergey Popov
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Galina Melnichenko
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Natalya Mokrysheva
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Marina Utkina
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| |
Collapse
|
41
|
Zhang Y, Yang Z, Liu Y, Pei J, Li R, Yang Y. Targeting lipid metabolism: novel insights and therapeutic advances in pancreatic cancer treatment. Lipids Health Dis 2025; 24:12. [PMID: 39806478 PMCID: PMC11727729 DOI: 10.1186/s12944-024-02426-0] [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: 11/11/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Lipid metabolism in cancer is characterized by dysregulated lipid regulation and utilization, critical for promoting tumor growth, survival, and resistance to therapy. Pancreatic cancer (PC) is a highly aggressive malignancy of the gastrointestinal tract that has a dismal 5-year survival rate of less than 10%. Given the essential function of the pancreas in digestion, cancer progression severely disrupts its function. Standard treatments for PC such as surgical resection, chemotherapy, and radiotherapy. However, these therapies often face significant challenges, including biochemical recurrence and drug resistance.Given these limitations, new therapeutic approaches are being developed to target tumor metabolism. Dysregulation of cholesterol biosynthesis and alterations in fatty acids (FAs), such as palmitate, stearate, omega-3, and omega-6, have been observed in pancreatic cancer. These lipids serve as energy sources, signaling molecules, and essential components of cell membranes. Their accumulation fosters an immunosuppressive tumor microenvironment that supports cancer cell proliferation and metastasis.Moreover, lipid metabolism dysregulation within immune cells, particularly T cells, impairs immune surveillance and weakens the body's defenses against cancer. Abnormal lipid metabolism also contributes to drug resistance in PC. Despite these challenges, targeting lipid metabolism may offer a promising therapeutic strategy. By enhancing lipid peroxidation, the induction of ferroptosis-a form of regulated cell death-could impair the survival of PC cells and hinder disease progression.
Collapse
Affiliation(s)
- Yanyan Zhang
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, Qinling, Hanzhong, 723001, China
| | - Zhichao Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Dalian Medical University, Dalian, China
| | - Yuchen Liu
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, Qinling, Hanzhong, 723001, China
| | - Jinjin Pei
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, Qinling, Hanzhong, 723001, China
| | - Ruojie Li
- Interventional Therapy Department, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, P.R. China.
| | - Yanhui Yang
- Emergency surgery Dapartment (Trauma center), The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, Henan, China.
| |
Collapse
|
42
|
Sun S, Jing X, Tong G, Chen C, Xie S, Wang C, Chen D, Zhao J, Qi Y, Zhang W, Liu C, Zhang G, Zhang J, Sun B, Wang Y, Lv Y. Loss of DDX24 inhibits lung cancer progression by stimulating IKBKG splicing-mediated autophagy. Theranostics 2025; 15:1879-1895. [PMID: 39897555 PMCID: PMC11780526 DOI: 10.7150/thno.102425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/21/2024] [Indexed: 02/04/2025] Open
Abstract
Rationale: Lung cancer remains a major global health burden with limited therapeutic options. Alternative splicing, a critical post-transcriptional process, contributes to lung cancer progression through autophagy, although the underlying mechanisms remain largely unexplored. This study aims to elucidate the role of DDX24 as a splicing factor that contributes to lung cancer progression via autophagy. Methods: To establish the link between DDX24 and lung cancer progression, we performed colony formation assays, growth curve analyses, and xenograft tumor models in nude mice. Mass spectrometry and RNA sequencing were employed to investigate the involvement of DDX24 in alternative splicing, with a specific focus on the splicing of IKBKG. The mechanisms by which DDX24 regulates autophagy were further explored using co-immunoprecipitation and luciferase reporter assays. Results: The splicing factor DDX24 is significantly elevated in lung cancer tissues. Loss of DDX24 suppresses lung cancer growth by promoting autophagy. We identified DDX24 as a splicing factor that plays critical roles in the regulation of alternative splicing. Mechanistically, DDX24 regulates the alternative splicing of autophagy-related genes, including IKBKG. We demonstrate that DDX24 directly binds to IKBKG pre-mRNA, whereas DDX24 ablation stimulates the generation of the long splicing isoform of IKBKG, thereby promoting autophagy through activating of the NF-kB signaling pathway and the transcription of the BECN1 gene. Functional rescue experiments confirm that the long IKBKG isoform-mediated autophagy confers the anti-tumor effects of DDX24 depletion. In addition, IKBKG-L is positively associated with improved survival in lung cancer patients. Conclusions: This study uncovers a novel regulatory axis involving DDX24, IKBKG splicing, and autophagy in lung cancer. Our findings suggest that targeting DDX24 may represent a promising therapeutic strategy for lung cancer treatment, offering new insights into the molecular underpinnings of this disease.
Collapse
Affiliation(s)
- Siwen Sun
- Department of Oncology & Sino-US Research Center for Cancer Translational Medicine, the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116023, China
| | - Xiaomeng Jing
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116023, China
| | - Guangquan Tong
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Chaoqun Chen
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116023, China
| | - Shuaijun Xie
- Department of Pathology, the First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Chong Wang
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116023, China
| | - Dan Chen
- Department of Pathology, the First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Jinyao Zhao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Yangfan Qi
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Wenjing Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Congcong Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Ge Zhang
- Department of Immunology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Jinrui Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Bing Sun
- Department of Thoracic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Yang Wang
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116023, China
| | - Yuesheng Lv
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University & Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116023, China
| |
Collapse
|
43
|
Purohit S, Mandal G, Biswas S, Dalui S, Gupta A, Chowdhury SR, Bhattacharyya A. AXL/GAS6 signaling governs differentiation of tumor-associated macrophages in breast cancer. Exp Cell Res 2025; 444:114324. [PMID: 39510154 DOI: 10.1016/j.yexcr.2024.114324] [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: 04/28/2024] [Revised: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Most epithelial cancers are infiltrated by prognostically relevant myelomonocytic cells. Immunosuppressive tumor associated macrophages (TAMs) and their precursor monocytic myeloid-derived suppressor cells (MDSCs) have previously been associated with worse outcomes in human breast cancer (BCa), yet the mechanism of immunosuppressive TAMs-polarization from myelomonocytic precursors is not completely understood. In this study, we show that persuaded AXL/GAS6 pathway alters macrophage phenotype from HLA-DRhighCD206lowCD163low classical phagocytic into HLA-DRlowCD206highCD163high immunosuppressive ones with accelerated BCa progression, and increased angiogenesis signature and invasion ability of cancer cells at tumor beds. Notably, both AXL and GAS6 expressions are upregulated in human invasive breast carcinoma, with maximum expression in triple negative histology type. Mechanistically, we demonstrate that AXL/GAS6 signaling drives immunosuppression by governing increased immunosuppressive IL10 production while dampening IL-1β expression within the tumor microenvironment (TME) of BCa. Further, AXL/GAS6 signaling promotes angiogenesis through the activation of PI3K/AKT and NF-κB signaling pathways. Our results unveil role of AXL/GAS6 axis in the differentiation of TAMs, which governs malignant growth, and suggest that therapies that uncouple AXL/GAS6 axis may exhibit therapeutic opportunity for otherwise undruggable Triple Negative Breast Cancer (TNBC) patients.
Collapse
Affiliation(s)
- Suman Purohit
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India; Department of Zoology, Gurudas College, 1/1, Suren Sarkar Road, Phool Bagan, Kolkata, 700054, West Bengal, India
| | - Gunjan Mandal
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India; Division of Cancer Biology, DBT-Institute of Life Sciences, Bhubaneswar, 751023, India
| | - Subir Biswas
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India; Tumor Immunology and Immunotherapy, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, Maharashtra, India
| | - Shauryabrota Dalui
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India
| | - Arnab Gupta
- Department of Surgical Oncology, Saroj Gupta Cancer Centre and Research Institute, Mahatma Gandhi Road, Kolkata, 700063, West Bengal, India
| | - Sougata Roy Chowdhury
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India; Translational Immunology Laboratory, Department of Life Science and Biotechnology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India.
| |
Collapse
|
44
|
Zhang X, Zhang M, Sun H, Wang X, Wang X, Sheng W, Xu M. The role of transcription factors in the crosstalk between cancer-associated fibroblasts and tumor cells. J Adv Res 2025; 67:121-132. [PMID: 38309692 PMCID: PMC11725164 DOI: 10.1016/j.jare.2024.01.033] [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: 11/29/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Transcription factors (TFs) fulfill a critical role in the formation and maintenance of different cell types during the developmental process as well as disease. It is believed that cancer-associated fibroblasts (CAFs) are activation status of tissue-resident fibroblasts or derived from form other cell types via transdifferentiation or dedifferentiation. Despite a subgroup of CAFs exhibit anti-cancer effects, most of them are reported to exert effects on tumor progression, further indicating their heterogeneous origin. AIM OF REVIEW This review aimed to summarize and review the roles of TFs in the reciprocal crosstalk between CAFs and tumor cells, discuss the emerging mechanisms, and their roles in cell-fate decision, cellular reprogramming and advancing our understanding of the gene regulatory networks over the period of cancer initiation and progression. KEY SCIENTIFIC CONCEPTS OF REVIEW This manuscript delves into the key contributory factors of TFs that are involved in activating CAFs and maintaining their unique states. Additionally, it explores how TFs play a pivotal and multifaceted role in the reciprocal crosstalk between CAFs and tumor cells. This includes their involvement in processes such as epithelial-mesenchymal transition (EMT), proliferation, invasion, and metastasis, as well as metabolic reprogramming. TFs also have a role in constructing an immunosuppressive microenvironment, inducing resistance to radiation and chemotherapy, facilitating angiogenesis, and even 'educating' CAFs to support the malignancies of tumor cells. Furthermore, this manuscript delves into the current status of TF-targeted therapy and considers the future directions of TFs in conjunction with anti-CAFs therapies to address the challenges in clinical cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xu Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xin Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| |
Collapse
|
45
|
Malik MNH, Ali S, Ali A, Alanzi AR, Atif M, Alharbi HA, Wang B, Raza M, Maqbool T, Anjum I, Jahan S, Alshammari SO, Solre GFB. Citronellol Induces Apoptosis via Differential Regulation of Caspase-3, NF-κB, and JAK2 Signaling Pathways in Glioblastoma Cell Line. Food Sci Nutr 2025; 13:e4678. [PMID: 39803280 PMCID: PMC11717069 DOI: 10.1002/fsn3.4678] [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: 08/10/2024] [Revised: 11/23/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
Citronellol (CT) is a naturally occurring lipophilic monoterpenoid which has shown anticancer effects in numerous cancerous cell lines. This study was, therefore, designed to examine CT's potential as an anticancer agent against glioblastoma (GBM). Network pharmacology analysis was employed to identify potential anticancer targets of CT. A comprehensive data mining was carried out to assess CT and GBM-associated target genes. Protein-protein interaction network was constructed to identify hub genes and later GO and KEGG enrichment analysis was performed to elucidate the possible mechanism. Human glioblastoma cell line "SF767" was used to confirm in silico findings. MTT, crystal violet, and trypan blue assays were performed to assess the cytotoxic effects of various concentrations of CT. Subsequently, ELISA and qPCR were performed to analyze the effects of CT on proapoptotic and inflammatory mediators. In silico findings indicated that CT differentially regulated proapoptotic and inflammatory pathways by activating caspase-3 and 8 and inhibiting nuclear factor-kappa B (NF-κB), tumor necrosis factor-α, Janus kinase 2 (JAK2). Molecular docking also demonstrated strong binding affinities of CT with the above-mentioned mediators when compared to 5-fluorouracil or temozolomide. In SF767 cell line, CT displayed dose-dependent cytotoxic and antioxidant effects, and upregulation of annexin-V, caspase-3, and 8 along with downregulation of inflammatory modulators. In a nutshell, it can be concluded from these findings that CT possesses robust anticancer activity which is mediated via differential regulation of caspase-3, JAK2, and NF-κB pathways.
Collapse
Affiliation(s)
| | - Sufyan Ali
- Faculty of PharmacyThe University of LahoreLahorePakistan
| | - Amir Ali
- Faculty of PharmacyThe University of LahoreLahorePakistan
| | - Abdullah R. Alanzi
- Department of Pharmacognosy, College of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Muhammad Atif
- Faculty of PharmacyThe University of LahoreLahorePakistan
| | - Hattan A. Alharbi
- Department of Pharmacognosy, College of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Bowen Wang
- College of Chinese MedicineHubei University of Chinese MedicineWuhanHubeiChina
| | - Moosa Raza
- Faculty of PharmacyThe University of LahoreLahorePakistan
| | - Tahir Maqbool
- Institute of Molecular Biology and Biotechnology (IMBB)The University of LahoreLahorePakistan
| | - Irfan Anjum
- Shifa College of Pharmaceutical SciencesShifa Tameer‐e‐Millat UniversityIslamabadPakistan
| | - Shah Jahan
- Department of ImmunologyUniversity of Health SciencesLahorePakistan
| | - Saud O. Alshammari
- Department of Pharmacognosy and Alternative Medicine, College of PharmacyNorthern Border UniversityRafhaSaudi Arabia
| | - Gideon F. B. Solre
- Department of Chemistry, Thomas J. R. Faulkner College of Science and TechnologyUniversity of LiberiaMonroviaMontserrado CountyLiberia
| |
Collapse
|
46
|
Lin J, Yuan M, Shi HY, Liu Q, Du S, Zhang MX, Li QQ, Yang ZB, Lin P. Phellinus linteus (Agaricomycetes) Polysaccharides Ameliorate Inflammatory Injury in H2O2-Induced Caco-2 Cells and DSS-Induced Ulcerative Colitis Mice. Int J Med Mushrooms 2025; 27:17-32. [PMID: 40094337 DOI: 10.1615/intjmedmushrooms.2025058082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Phellinus linteus (Agaricomycetes) is a valuable medicinal mushroom traditionally used as a food supplement and medicinal ingredient. Polysaccharides of Ph. linteus (PLP) possess strong anti-inflammatory effects and gut microbiota modulating properties. However, the mechanism of its efficacy in ulcerative colitis (UC) remains unclear. This study utilized 1mM H2O2 to induce an in vitro model of UC in Caco-2 cells. Additionally, a 3% solution of dextran sulfate sodium salt (DSS) was employed to establish an in vivo UC model in mice. After treating the cells with PLP at various concentrations, there was a significant reduction in the mRNA expression of TNF-α and IL-6, and the nuclear factor-κB (NF-κB) signaling pathway was also inhibited. Concurrently, symptoms such as colon shortening, weight loss, and a decrease in disease activity index (DAI) scores were significantly improved in UC mice. Additionally, the treatment led to downregulated expression of TNF-α and IFN-γ mRNA in colon tissues. PLP had shown potential in reducing inflammation and oxidative stress in Caco-2 cells, demonstrating therapeutic effects in treating UC-like inflammation by inhibiting the NF-κ signaling pathway and activating the nuclear factor erythroid derived 2-like 2(Nrf2)/heme oxygenase-1(HO-1) signaling pathway. These findings suggest that PLP has great potential for further investigation and development in UC treatment.
Collapse
Affiliation(s)
- Jun Lin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P.R. China
| | - Meng Yuan
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P.R. China
| | - Hong-Yu Shi
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P.R. China
| | - Qiang Liu
- Shandong Focusfreda Biotech Co. Ltd., Qufu 273165, P.R. China
| | - Shuai Du
- Shandong Focusfreda Biotech Co. Ltd., Qufu 273165, P.R. China
| | - Mei-Xia Zhang
- Shandong Focusfreda Biotech Co. Ltd., Qufu 273165, P.R. China
| | - Qu-Quan Li
- Shandong Focusfreda Biotech Co. Ltd., Qufu 273165, P.R. China
| | - Zhen-Bang Yang
- Shandong Focusfreda Biotech Co. Ltd., Qufu 273165, P.R. China
| | - Pei Lin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
47
|
Zhong X, Zhang W, Zhang W, Yu N, Li W, Song X. FASN contributes to ADM resistance of diffuse large B-cell lymphoma by inhibiting ferroptosis via nf-κB/STAT3/GPX4 axis. Cancer Biol Ther 2024; 25:2403197. [PMID: 39345091 PMCID: PMC11445901 DOI: 10.1080/15384047.2024.2403197] [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: 01/04/2024] [Revised: 08/06/2024] [Accepted: 09/07/2024] [Indexed: 10/01/2024] Open
Abstract
Drug resistance is a critical impediment to efficient therapy of diffuse large B-cell lymphoma (DLBCL) patients. Recent studies have highlighted the association between ferroptosis and drug resistance that has been reported. Fatty acid synthase (FASN) is always related to a poor prognosis. In this study, we investigate the impact of FASN on drug resistance in DLBCL and explore its potential modulation of ferroptosis mechanisms. The clinical correlation of FASN mRNA expression was first analyzed to confirm the role of FASN on drug resistance in DLBCL based on the TCGA database. Next, the impact of FASN on ferroptosis was investigated in vitro and in vivo. Furthermore, a combination of RNA-seq, western blot, luciferase reporter, and ChIP experiments was employed to elucidate the underlying mechanism. The prognosis for patients with DLBCL was worse when FASN was highly expressed, particularly in those undergoing chemotherapy for Adriamycin (ADM). FASN promoted tumor growth and resistance of DLBCL to ADM, both in vitro and in vivo. It is noteworthy that this effect was achieved by inhibiting ferroptosis, since Fer-1 (a ferroptosis inhibitor) treatment significantly recovered the effects of silencing FASN on inhibiting ferroptosis, while Erastin (a ferroptosis inducer) treatment attenuated the impact of overexpressing FASN. Mechanistically, FASN activated NF-κB/STAT3 signaling pathway through phosphorylating the upstream IKKα and IκBα, and the activated STAT3 promoted GPX4 expression by directly binding to GPX4 promoter. FASN inhibits ferroptosis in DLBCL via NF-κB/STAT3/GPX4 signaling pathway, indicating its critical role in mediating ADM resistance of DLBCL.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Cell Line, Tumor
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Drug Resistance, Neoplasm
- Fatty Acid Synthase, Type I/metabolism
- Fatty Acid Synthase, Type I/genetics
- Ferroptosis/drug effects
- Ferroptosis/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Mice, Nude
- NF-kappa B/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
- Prognosis
- Signal Transduction/drug effects
- STAT3 Transcription Factor/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Xing Zhong
- Departments of Lymphatic and Hematological Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, P. R. China
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, P. R. China
| | - Weiwei Zhang
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, P. R. China
- Nanchang Medical College, Nanchang, Jiangxi, P. R. China
| | - Weiming Zhang
- Nanchang Medical College, Nanchang, Jiangxi, P. R. China
| | - Nasha Yu
- Departments of Lymphatic and Hematological Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, P. R. China
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, P. R. China
| | - Wuping Li
- Departments of Lymphatic and Hematological Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, P. R. China
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, P. R. China
| | - Xiangxiang Song
- Departments of Lymphatic and Hematological Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, P. R. China
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, P. R. China
| |
Collapse
|
48
|
Song Z, Chen H, Wang X, Zhang Z, Li H, Zhao H, Liu Y, Han Q, Zhang J. Napabucasin-loaded PLGA nanoparticles trigger anti-HCC immune responses by metabolic reprogramming of tumor-associated macrophages. J Transl Med 2024; 22:1125. [PMID: 39707412 DOI: 10.1186/s12967-024-05917-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND JAK/STAT3 is one of the critical signaling pathways involved in the occurrence and development of hepatocellular carcinoma (HCC). BBI608 (Napabucasin), as a novel small molecule inhibitor of STAT3, has shown previously excellent anti-HCC effects in vitro and in mouse models. However, low bioavailability, high cytotoxicity and other shortcomings limit its clinical application. In this study, PLGA was selected to prepare Napabucasin PLGA nanoparticles (NPs) by solvent evaporation method, overcoming these limitations and improving the passive targeting effect that nanoparticle mediated. Base on this, we systematically evaluated the anti-HCC effect of Napabucasin-PLGA NPs and explored the underlying mechanisms. METHODS Napabucasin-PLGA NPs were prepared by solvent evaporation method. CCK-8 assay, Annexin V/PI double staining, RT-qPCR, colony formation assay, and Western blotting were performed to evaluate the anti-HCC effect of Napabucasin-PLGA NPs in vitro. Proliferation assay and migration assay were used to detect the effects of Napabucasin-PLGA NPs-treated HCC-TAMs on tumor biological characteristics of HCC cells. Flow cytometry was used to detect anti-HCC immune responses induced by Napabucasin-PLGA NPs in vivo. RESULTS Our results demonstrated that Napabucasin-PLGA NPs could improve the bioavailability of Napabucasin and enhance Napabucasin-mediated the anti-HCC effects in vitro and in vivo with no significant drug toxicity. In addition to the direct inhibitory effects on the tumor biological characteristics of HCC cells, Napabucasin-PLGA NPs could promote the polarization of macrophages from tumor-promoting M2-type to anti-tumor M1-type, improving the tumor immune microenvironment and augmenting T cell-mediated anti-tumor responses. The underlining mechanisms showed Napabucasin-PLGA NPs suppressed the STAT3/FAO signaling axis in HCC-induced tumor-associated macrophages (TAMs). CONCLUSIONS These findings demonstrated Napabucasin-PLGA NPs is a potential therapeutic candidate for HCC, and provided a new theoretical and experimental basis for further development and clinical application of Napabucasin.
Collapse
Affiliation(s)
- Zhenwei Song
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Hongfei Chen
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xueyao Wang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Hui Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yang Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
49
|
Mercuţ R, Ciurea ME, Traşcă ET, Ionescu M, Mercuţ MF, Rădulescu PM, Călăraşu C, Streba L, Ionescu AG, Rădulescu D. Applying Neural Networks to Analyse Inflammatory, Sociodemographic, and Psychological Factors in Non-Melanoma Skin Cancer and Colon Cancer: A Statistical and Artificial Intelligence Approach. Diagnostics (Basel) 2024; 14:2759. [PMID: 39682667 DOI: 10.3390/diagnostics14232759] [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: 11/02/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Chronic inflammation and psychosocial factors significantly influence cancer progression and patient behavior in seeking medical care. Understanding their interplay is essential for enhancing early detection and developing personalized treatment strategies. This study aims to develop a comprehensive patient profiling model by comparing non-melanoma skin cancer (NMSC) and colorectal cancer (CRC). The goal is to identify common and distinct patterns in inflammation and psychosocial factors that affect disease progression and clinical presentation. Methods: We conducted a comparative analysis of patients diagnosed with NMSC and CRC, integrating clinical data with sociodemographic and psychological assessments. Advanced neural network algorithms were employed to detect subtle patterns and interactions among these factors. Based on the analysis, a cancer risk assessment questionnaire was developed to stratify patients into low-, moderate-, and high-risk categories. Results: Patients with low systemic inflammation and adequate vagal tone, supported by a stable family environment, demonstrated heightened sensitivity to subclinical symptoms, enabling earlier diagnosis and timely intervention. Conversely, patients with high systemic inflammation and reduced vagal tone, often influenced by chronic stress and unstable family environments, presented at more advanced disease stages. The developed risk assessment tool effectively classified patients into distinct risk categories, facilitating targeted preventive measures and personalized therapeutic strategies. Neural network profiling revealed significant interactions between biological and psychosocial factors, enhancing our understanding of their combined impact on cancer progression. Conclusions: The integrated profiling approach and the newly developed risk assessment questionnaire have the potential to transform cancer management by improving early detection, personalizing treatment strategies, and addressing psychosocial factors. This model not only enhances clinical outcomes and patient quality of life but also offers a framework adaptable to other cancer types, promoting a holistic and patient-centered approach in oncology.
Collapse
Affiliation(s)
- Răzvan Mercuţ
- Department of Plastic and Reconstructive Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Marius Eugen Ciurea
- Department of Plastic and Reconstructive Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Emil Tiberius Traşcă
- The Surgery Clinic of "Dr. Ștefan Odobleja Emergency Military Hospital", General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Mihaela Ionescu
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Maria Filoftea Mercuţ
- Department of Ophthalmology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | | | - Cristina Călăraşu
- Department of Pneumology, University of Pharmacy and Medicine Craiova, 200349 Craiova, Romania
| | - Liliana Streba
- Department of Oncology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Alin Gabriel Ionescu
- Department of Medical History, University of Pharmacy and Medicine Craiova, 200349 Craiova, Romania
| | - Dumitru Rădulescu
- The Surgery Clinic of "Dr. Ștefan Odobleja Emergency Military Hospital", General Surgery Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
50
|
Pan Y, Ma T, Chen D, Wang Y, Peng Y, Lu T, Yin X, Li H, Zhang G, Wang X. Scutellaria barbata D.Don and Scleromitrion diffusum (Willd.) R.J.Wang inhibits the progression of triple negative breast cancer though the activation inhibition of NF-κB triggered by CAFs-derived IL6. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118656. [PMID: 39121924 DOI: 10.1016/j.jep.2024.118656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The treatment options for triple-negative breast cancer (TNBC) are limited. Traditional Chinese Medicine (TCM) plays an important role in the treatment of TNBC. The herb pair Scutellaria barbata D.Don and Scleromitrion diffusum (Willd.) R.J.Wang (SH) is commonly used in clinical practice for its anti-tumor properties. It has been proven to have good therapeutic effects on tumor-related diseases, but the underlying molecular mechanisms are not yet fully explained. AIM OF STUDY Through bioinformatics, it was validated that IL6, primarily derived from cancer-associated fibroblasts (CAFs), is associated with poor prognosis. Additionally, cell and animal experiments confirmed that SH inhibits tumor proliferation, migration, and growth in an orthotopic tumor model by suppressing the IL6/NF-κB pathway. MATERIALS AND METHODS GEO, TCGA and HPA databases were used to analyze the prognostic value of CAFs and IL6, then IL6 resource was detected. After the bioinformatics, the influence of CAFs and CAFs-derived IL6 on TNBC was verified by experiments both in vitro and in vivo. Cell clone formation assay, wound-Healing assay, and Transwell assay were used to detect the promotion of CAFs and CAFs-derived IL6 and the inhibition of SH in vitro. TNBC model in mice was used to prove the promotion of CAFs and CAFs-derived IL6 and the inhibition of SH in vivo. The biological pathway of NF-κB was explored by western blotting through detecting unique molecules. RESULTS Bioinformatics analysis revealed that higher proportion of CAFs and elevated level of IL6 were significantly associated with poor prognosis in TNBC. At the same time, IL6 was proved predominantly derived from CAFs. After the indication of bioinformatics, experiments in vitro demonstrated that both CAFs and IL6 could enhance the clone formation and migration ability of MDA-MD-231 cells (231), furthermore, the promotion of CAFs was related with the level of IL6. Based on these data, mechanism was detected that CAFs-derived IL6 enhancement was closely related to the activation of NF-κB signaling pathway, while the activation can be reduced by SH. In the end, the promotion of CAFs/CAFs-derived IL6/NF-κB and the efficacy of SH inhibition were both confirmed by experiments in vivo. CONCLUSIONS Bioinformatics data indicates that higher proportion of CAFs and higher level of CAFs-derived IL6 are significantly related to poorer survival of TNBC. CAFs and CAFs-derived IL6 were proved to promote the progression of TNBC both in vitro and in vivo, and the process of which was significantly related to the activation of NF-κB. SH inhibited the progress of TNBC, which was proved to be closely related to CAFs/CAFs-derived IL6/NF-κB.
Collapse
Affiliation(s)
- Yuancan Pan
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Tingting Ma
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Dong Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Yue Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu Peng
- Shandong University of Traditional Chinese Medicine, Shandong, 250355, China
| | - Taicheng Lu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Xiaohui Yin
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Haiming Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Ganlin Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Xiaomin Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| |
Collapse
|