1
|
Luo H, Zhang Y, Yao Y, Zhang Q, Lin S, Wang S, Wang L, Lu G, Lin Z, Lin D. A novel galactoglucan from Ganoderma lucidum ameliorates ethanol-induced gastric ulcers by modulating FAK-MAPK signaling pathway. Carbohydr Polym 2025; 360:123594. [PMID: 40399003 DOI: 10.1016/j.carbpol.2025.123594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 05/23/2025]
Abstract
Gastric ulcer (GU) is a prevalent gastrointestinal disorder that significantly impacts patients' quality of life. Current treatments, including proton pump inhibitors and H2 receptor antagonists, often present limitations such as adverse side effects and incomplete healing, highlighting the urgent need for new therapeutic agents. Ganoderma lucidum polysaccharide peptides (GL-PP) have gained attention for their potential gastroprotective properties. Notably, we characterized a novel GL-PP named GL-PPQ3, assessing its efficacy in alleviating GUs using an ethanol-induced GU mouse. Our findings indicated that GL-PPQ3 is a hyperbranched galactoglucan with a molecular weight of 41.08 kDa, predominantly composed of glucose and galactose. Comprehensive analysis, including methylation and one-dimensional and two-dimensional nuclear magnetic resonance (NMR), elucidated its structural composition, revealing the presence of 1,6-α-D-Galp, 1,2,6-α-D-Galp, 1,6-β-D-Glcp, 1,3,6-β-D-Glcp, and 1,4-β-D-Glcp residues. X-ray diffraction (XRD) confirmed a predominantly amorphous structure and scanning electron microscopy (SEM) displayed aggregated spherical formations. Mechanistically, GL-PPQ3 exhibited its Gastroprotective effects through the regulation of the FAK-mediated MAPK signaling pathway. This study underscores the therapeutic potential of GL-PPQ3 as a promising candidate for GU treatment, paving the way for developing novel functional agents derived from natural sources.
Collapse
Affiliation(s)
- Hongjian Luo
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Yukun Zhang
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing 404120, China; State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yang Yao
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing 404120, China
| | - Qiaoling Zhang
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing 404120, China
| | - Shuqian Lin
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Saizhen Wang
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Lianfu Wang
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Guodong Lu
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China; College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China.
| | - Zhanxi Lin
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China.
| | - Dongmei Lin
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China.
| |
Collapse
|
2
|
Ertilav K, Nazıroğlu M. Cyclophosphamide Induces Glioblastoma Tumor Cell Death and Oxidative Stress Through the Increase of TRPM2 Channel Stimulation: The Role of Carvacrol. Cell Biol Int 2025. [PMID: 40401344 DOI: 10.1002/cbin.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Cyclophosphamide (CP) damages glioblastoma cells by producing an excessive amount of intracellular (iROS) and mitochondrial (mROS) reactive oxygen species. Both iROS and mROS are produced when TRPM2 is activated, but they are decreased when carvacrol (CAR) and N-(p-amylcinnamoyl) anthranilic acid (ACA) inhibit it. Therefore, iROS, and mROS via upregulating Ca2+ influx and apoptosis in glioblastoma (DBTRG-05MG) cells, CP-mediated TRPM2 stimulation may cause oxidant and apoptotic activities. We investigated how TRPM2 activation not only promotes DBTRG-05MG death but also modifies oxidative damage and apoptosis to counteract the effects of ACA and CAR. The groups of control (CN), CAR (200 μM for 24 h), CP (2 mM for 24 h), and CP + CAR were induced in the DBTRG-05MG. While cytosolic free Ca2+ levels decreased in the cells as a result of the CAR and ACA treatments, they were further elevated in the CP group by the stimulation of TRPM2 (H2O2). The cells in the CP group had higher levels of dead cell percentage, apoptosis, mitochondrial membrane dysfunction, mROS, iROS, and caspases -3, -8, and -9 than the CN and CAR cells, although their levels were lower in the CP + CAR than in the CP only. CAR incubation increased the CP-induced glutathione concentration and cell viability percentage declines. In summary, the anticancer effect of CP was enhanced by TRPM2 stimulation, while CP-induced oxidative stress and DBTRG-05MG death were reduced by TRPM2 suppression when CAR was treated. TRPM2 activation may be a possible tumor killer channel due to oxidative glioma damage caused by CP.
Collapse
Affiliation(s)
- Kemal Ertilav
- Department of Neurosurgery, School of Medicine, University of Suleyman Demirel, Isparta, Türkiye
| | - Mustafa Nazıroğlu
- Neuroscience Application and Research Center (NOROBAM), University of Suleyman Demirel, Isparta, Türkiye
- BSN Health, Analyses, Innov., Consult., Org., Agricul. Ltd., Isparta, Türkiye
- Department of Biophysics, School of Medicine, University of Suleyman Demirel, Isparta, Türkiye
| |
Collapse
|
3
|
Gong Y, Zhang W, Wang X, Zheng S, Liu H, Lin Q, Wang M, Ma J, Zhang Y, Li T, Hu Y. A previously uncharacterized role of TAp73 in ferroptosis by modulating oxidative homeostasis in cervical cancer. Cancer Lett 2025:217815. [PMID: 40409453 DOI: 10.1016/j.canlet.2025.217815] [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: 04/07/2025] [Revised: 05/17/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
While the tumor-suppressive functions of p53 are well established, the role of its homolog, TAp73, in cancer remains incompletely characterized and is a subject of active investigation. In this study, we observed downregulation of TAp73 protein expression in cervical cancer tissues, which significantly correlated with adverse clinical outcomes. Through co-expression network analysis, we identified functional associations between TAp73 and key pathways involved in lipid metabolism and redox homeostasis-both critical regulators of ferroptosis, an iron-dependent form of programmed cell death mediated by lipid peroxidation. Mechanistically, we demonstrate that TAp73 promotes ferroptosis by directly upregulating the transcription of β-transducin repeat-containing protein (β-TRCP), thereby facilitating the ubiquitin-dependent degradation of nuclear factor erythroid 2-related factor 2 (NRF2), a master regulator of cellular antioxidant defenses. This TAp73-mediated suppression of NRF2 activity renders cells more susceptible to ferroptotic death. Furthermore, TAp73 expression is transcriptionally induced during ferroptosis through the combined inactivation of enhancer of zeste homolog 2 (EZH2), a core component of polycomb repressive complex 2, and activation of E2F transcription factor 1 (E2F1). Notably, pharmacological inhibition of EZH2 synergized with sulfasalazine (SAS) to enhance ferroptosis in vivo, an effect largely dependent on TAp73. Together, these findings delineate a novel ferroptosis regulatory axis-EZH2/TAp73/β-TRCP/NRF2-and highlight its potential as a therapeutic target for cervical cancer intervention.
Collapse
Affiliation(s)
- Yafan Gong
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Wenxin Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Shanliang Zheng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Hao Liu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Qingyu Lin
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Meiqi Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001
| | - Jiangwen Ma
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001
| | - Yi Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Tianyu Li
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000.
| |
Collapse
|
4
|
Wolf F, Dietrich-Ntoukas T, Reinach PS, Pleyer U, Mergler S. Nerve Growth Factor Modulates Regulatory Cell Volume Behavior via Stimulating TRPV1, TRPM8 Channels and Inducing Ca 2+ Signaling in Human Conjunctival Epithelial Cells. Cells 2025; 14:719. [PMID: 40422222 DOI: 10.3390/cells14100719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/28/2025] Open
Abstract
NGF plays important roles in ocular surface homeostasis and different pathological conditions. One effect includes promoting conjunctival epithelial cell differentiation and mucin secretion. This study characterizes the individual roles of TRPV1 and TRPM8 channel activity in mediating the effects of NGF on intracellular Ca2+ regulation and its alteration of regulatory cell volume responses to anisosmotic challenges in human conjunctival epithelial cells (IOBA-NHC). With fura-2/AM-loaded cells, the effects of 40 µM capsaicin and 20 µM AMG 9810 on Ca2+ regulation confirm functional TRPV1 expression. TRPM8 expression is evident since 500 µM menthol and 20 µM AMTB have opposing effects on [Ca2+]i. AMG 9810 and AMTB (both 20 µM) suppress the responses to NGF (100 ng/mL). With calcein/AM-loaded cells, the effects of these mediators are evaluated on apparent cell volume responses induced by an anisosmotic challenge. NGF decreases the apparent cell volume that AMG 9810 suppresses, whereas AMTB (both 20 µM) augments this response. Therefore, NGF interacts with TRPV1 and TRPM8 to induce opposing effects on cell volume regulatory behavior. These opposing effects suggest that the signaling pathways and effectors that mediate responses to TRPV1 and TRPM8 activation are not the same.
Collapse
Affiliation(s)
- Friedrich Wolf
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tina Dietrich-Ntoukas
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Peter S Reinach
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325015, China
| | - Uwe Pleyer
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Stefan Mergler
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
5
|
Wang S, Xu D, Wang Y, Zhou Y, Xiao L, Li F, Tu J, Qin W, Tian S, Zheng B, Wang Y, Yuan XL, Liu Y, Liu B. A Bifunctional Antibody Targeting PD-1 and TGF-β Signaling Has Antitumor Activity in Combination with Radiotherapy and Attenuates Radiation-Induced Lung Injury. Cancer Immunol Res 2025; 13:767-784. [PMID: 39878763 PMCID: PMC12046334 DOI: 10.1158/2326-6066.cir-23-0903] [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: 10/27/2023] [Revised: 04/03/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Radio-immunotherapy has antitumor activity but also causes toxicity, which limits its clinical application. JS-201 is a dual antibody targeting PD-1 and TGF-β signaling. We investigated the antitumor effect of JS-201 combined with radiotherapy (RT) and the effect on radiation-induced lung injury (RILI). Different tumor models were established to detect the antitumor effects of the combination of JS-201 and RT, and RILI models were established to observe the effects of JS-201. Transcriptome sequencing showed that JS-201 optimized the tumor microenvironment by inhibiting extracellular matrix formation and angiogenesis. Combining JS-201 with RT further increased the inflammatory response and immune infiltration and showed great abscopal effects in Lewis lung cancer luciferase-positive models. Single-cell sequencing demonstrated that JS-201 reduced fibroblast proliferation by inhibiting the TGF-β/Smad pathway and the release of neutrophil extracellular traps mediated by ROS, thereby relieving radiation-induced pulmonary fibrosis. In conclusion, the JS-201 and RT combination enhances antitumor effects while mitigating acute and chronic RILI, and it may have potential for translational investigation as a cancer treatment strategy.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Duo Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuehua Zhou
- Top Alliance Biosciences Inc., Suzhou, China
| | - Lingyan Xiao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wan Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sidan Tian
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Bolong Zheng
- School of Computer Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yihua Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Xiang-lin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanhui Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Assimakopoulou M, Soufli K, Melachrinou M. Polycystins Expression in Astrocytic Gliomas. Biomedicines 2025; 13:884. [PMID: 40299470 PMCID: PMC12025129 DOI: 10.3390/biomedicines13040884] [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: 02/24/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Polycystin 1 (PC1) and polycystin 2 (PC2) proteins are members of the transient receptor potential (TRP) channels family and are encoded from PKD1 and PKD2 genes, respectively. Until recently, the role of PKD1 and PKD2 has been associated with the pathogenesis of the kidney since mutations in these genes cause autosomal dominant polycystic kidney disease (ADPKD). Recent data implicates polycystins in the pathogenesis of solid tumors. In this aspect, the expression of PKD1 and PKD2 in human astrocytomas is largely unknown. The aim of the present research study was to investigate the expression of PKD1 and PKD2 in astrocytic tumors and correlate it with clinicopathological characteristics such as the grade of malignancy, age, and gender of the patients. Methods: A total of 70 cases-corresponding to 8 grade II (diffuse fibrillary astrocytomas), 12 grade III (anaplastic astrocytomas), and 50 grade IV (glioblastomas multiforme)-were examined. The mRNA expression levels of PKD1 and PKD2 were determined through molecular qRT-PCR analysis using the relative quantification ΔΔCt method and the expression of PC1 and PC2 was detected through immunohistochemistry using the semi-quantitative H-score system. Results: Increased levels of PKD1 and PKD2 in astrocytomas were found compared with that of a normal brain (p < 0.05). Glioblastomas demonstrated the greatest increase in PKD1 and PKD2 expression compared to other grades of malignancy (p < 0.05). The same pattern of expression showed PC1 and PC2 proteins. A significant correlation between PKD1 and PKD2 as well as PC1 and PC2 expressions was found (p < 0.05). Although no association was detected between PC1 or PC2 and Ki67 expression (p > 0.05), a significant correlation between PC1 and p53 immunoexpressions, in grade III and between PC2 and p53 immunoexpressions, in grade II astrocytomas (p < 0.01) has emerged. PC1 expression was correlated with age of the patients (p < 0.05). PKD1 and PKD2 expression were negatively correlated with the prognosis of glioma patients. Conclusions: The results of this study indicate the potential involvement of polycystins in the pathogenesis of astrocytomas. However, further research is required to fully understand the mechanisms that these molecules are implicated.
Collapse
Affiliation(s)
- Martha Assimakopoulou
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Konstantina Soufli
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Maria Melachrinou
- Department of Pathology, School of Medicine, University of Patras, 26504 Patras, Greece;
| |
Collapse
|
7
|
Yu M, Fei B, Chu S. Targeting HNRNPA2B1 to overcome chemotherapy resistance in gastric cancer stem cells: Mechanisms and therapeutic potential. J Biol Chem 2025; 301:108234. [PMID: 39870196 PMCID: PMC11999277 DOI: 10.1016/j.jbc.2025.108234] [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: 07/16/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/29/2025] Open
Abstract
Gastric cancer (GC) remains a significant global health challenge, particularly due to the resistance of gastric cancer stem cells (GCSCs) to chemotherapy. This study investigates the role of heterogeneous nuclear ribonucleoprotein A2/B1 (HNRNPA2B1), a member of the heterogeneous nuclear ribonucleoproteins (hnRNPs), in modulating mitochondrial metabolic reprogramming and contributing to chemoresistance in GCSCs. Through extensive analysis of tumor cancer genome atlas (TCGA) and gene expression omnibus (GEO) datasets, HNRNPA2B1 was identified as a key regulator in GCSCs, correlating with poor prognosis and enhanced resistance to chemoresistance. CRISPR-Cas9 mediated knockout of HNRNPA2B1 in GCSCs led to a significant decrease in mitochondrial function, reduced migration, invasion, and sphere formation abilities, and markedly increased apoptosis. These changes were accompanied by a shift in metabolic activity, evidenced by decreased oxygen consumption and increased extracellular acidification. Our results highlight HNRNPA2B1 as a pivotal factor in sustaining the malignant phenotype of GCSCs and present it as a potential therapeutic target to improve chemotherapy efficacy in GC.
Collapse
Affiliation(s)
- Miao Yu
- Department of Gastrointestinal colorectal and anal surgery, The Third Bethune Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Bingyuan Fei
- Department of Gastrointestinal colorectal and anal surgery, The Third Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Songtao Chu
- Department of Forensic Medicine of Basic Medical College, Beihua University, Jilin, Jilin Province, China.
| |
Collapse
|
8
|
Feng H, Zhou K, Yuan Q, Liu Z, Zhang T, Chen H, Xu B, Sun Z, Han Z, Liu H, Yu S, Chen T, Li G, Zhou W, Yu J, Huang W, Jiang Y. Noninvasive Assessment of Vascular Endothelial Growth Factor and Prognosis in Gastric Cancer Through Radiomic Features. Clin Transl Gastroenterol 2025; 16:e00802. [PMID: 39787380 PMCID: PMC11932601 DOI: 10.14309/ctg.0000000000000802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION Gastric cancer (GC) is a leading cause of cancer-related deaths worldwide, with delayed diagnosis often limiting effective treatment options. This study introduces a novel, noninvasive radiomics-based approach using [18F] FDG PET/CT (fluorodeoxyglucose positron emission tomography/computed tomography) to predict vascular endothelial growth factor (VEGF) status and survival in patients with GC. The ability to noninvasively assess these parameters can significantly influence therapeutic decisions and outcomes. METHODS We conducted a retrospective study involving patients diagnosed with GC, stratified into training, validation, and test groups. Each patient underwent a [18F] FDG PET/CT scan, and radiomic features were extracted using dedicated software. A Radiomics Score (RS) was calculated, serving as a predictor for VEGF status. Statistical analyses included logistic regression and Cox proportional hazards models to evaluate the predictive power of RS on survival outcomes. RESULTS The developed radiomics model demonstrated high predictive accuracy, with the RS formula achieving an area under the receiver operating characteristic curve of 0.861 in the training cohort and 0.857 in the validation cohort for predicting VEGF status. The model also identified RS as an independent prognostic factor for survival, where higher RS values correlated with poorer survival rates. DISCUSSION The findings underscore the potential of [18F] FDG PET/CT radiomics in transforming the management of GC by providing a noninvasive means to assess tumor aggressiveness and prognosis through VEGF status. This model could facilitate earlier and more tailored therapeutic interventions, potentially improving survival outcomes in a disease marked by typically late diagnosis and limited treatment success.
Collapse
Affiliation(s)
- Hao Feng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kangneng Zhou
- College of Computer Science, Nankai University, Tianjin, China
| | - Qingyu Yuan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwei Liu
- Department of PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Taojun Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Benjamin Xu
- Lynbrook High School, San Jose, California, USA
| | - Zepang Sun
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhen Han
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Liu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shitong Yu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guoxin Li
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Wenlan Zhou
- Department of PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weicai Huang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuming Jiang
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
9
|
Zhou L, Zhou G, Li J, Guo R, Dong H. NCX1 interacts with TRPA1 to promote cell proliferation and tumor growth of colon cancer via disruption of calcium homeostasis. J Adv Res 2025:S2090-1232(25)00129-8. [PMID: 40010607 DOI: 10.1016/j.jare.2025.02.031] [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: 01/03/2025] [Revised: 02/20/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025] Open
Abstract
INTRODUCTION Aberrant Ca2+ signaling plays a critical role in the hallmark of cancer, but its regulatory mechanisms in tumorigenesis remain largely unclear. Na+/Ca2+ exchanger 1 (NCX1) functions as a bidirectional Na+ and Ca2+ transporter, operating in either Ca2+ entry or exit mode, while the transient receptor potential ankyrin 1 (TRPA1) serves as a Ca2+-permeable channel. Both play crucial roles in maintaining normal homeostasis of cytosolic Ca2+ ([Ca2+]cyt). Although each of them has been implicated in some tumorigenesis, the potential coordination between NCX1 and TRPA1 in the pathogenesis of colon cancer (CC) remains unexplored. OBJECTIVES We investigated the impact of NCX1- and TRPA1-mediated Ca2+ signaling on CC and the underlying mechanisms. METHODS The cell experiments were conducted using the human normal colonic epithelial cell line (HCoEpiC) and human colon cancer cell lines (Caco-2, SW620, and DLD-1). We performed stable transfection to knock down NCX1 or TRPA1 genes and employed CCK8, colony formation, and flow cytometry assays to assess cell proliferation. We employed RT-qPCR, Western blotting, immunofluorescence and co-immunoprecipitation assays to explore the expression and regulatory relationship between NCX1 and TRPA1. Calcium and sodium assays were used to determine [Ca2+]cyt and [Na+]cyt. Finally, we used the xenografted tumor model to verify their impact on CC development in vivo. RESULTS NCX1 and TRPA1 were parallelly over-expressed, co-localized, and bound, and their functional activities were enhanced in human CC cells. NCX1 functions in Ca2+ exit mode to expel [Ca2+]cyt, in which TRPA1 function was clearly verified as well. Moreover, when the Ca2+ exit mode of NCX1 was inhibited, TRPA1 activation resulted in a larger amount of [Ca2+]cyt to suppress cell proliferation through inhibiting ERK1/2 and β-catenin phosphorylation. NCX1 or TRPA1 knockdown significantly diminished tumor growth in vivo. CONCLUSION TRPA1 channels couple with the Ca2+ exit mode of NCX1 to maintain a moderate increase in [Ca2+]cyt in CC cells, thereby promoting CC cell proliferation and tumor growth through ERK1/2 and β-catenin phosphorylation. Consequently, the NCX1/TRPA1 coupling may serve as an innovative target for preventing and treating CC.
Collapse
Affiliation(s)
- Liyong Zhou
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China
| | - Guolong Zhou
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China
| | - Junhui Li
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China
| | - Ruihong Guo
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China.
| | - Hui Dong
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China.
| |
Collapse
|
10
|
Yang J, Luo Y, Yao Z, Wang Z, Jiang K. Theoretical perspectives and clinical applications of non-coding RNA in lung cancer metastasis: a systematic review. Discov Oncol 2025; 16:169. [PMID: 39937377 PMCID: PMC11822152 DOI: 10.1007/s12672-025-01919-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
Lung cancer is one of the deadliest malignancies worldwide, with distant metastasis being a major cause of death. However, the specific mechanisms of lung cancer metastasis remain unclear. NcRNAs, a widely present type of non-coding RNAs in the body, constitute about 98% of the human genome, lacking protein-coding capacity but involved in various cellular processes such as proliferation, apoptosis, invasion, and migration. Studies have shown that ncRNAs play a crucial role in the metastasis of lung cancer, although research in this area is limited. This review summarizes the biological origins and functions of ncRNAs, their specific roles and mechanisms in lung cancer metastasis, and discusses their potential for early screening and therapeutic applications in lung cancer. Furthermore, it outlines the challenges in translating basic advancements of ncRNAs in lung cancer metastasis into clinical practice.
Collapse
Affiliation(s)
- Jie Yang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China
| | - Yi Luo
- The Clinical Medical College, Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Zuhuan Yao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China
| | - Zhaokai Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China
| | - Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China.
| |
Collapse
|
11
|
Li CL, Yao ZY, Qu C, Shao GM, Liu YK, Pei XY, Cao JY, Wang ZS. Machine learning model reveals the risk, prognosis, and drug response of histamine-related signatures in pancreatic cancer. Discov Oncol 2025; 16:155. [PMID: 39934551 DOI: 10.1007/s12672-025-01910-y] [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: 11/18/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Histamine, a critical inflammatory mediator, is generated by both mast cells and specific tumor cells, and it plays a fundamental role in inflammatory and immune responses. In the current scientific landscape, histamine-related genes (HRGs) and their associated pathways have been validated to be implicated in the development and advancement of cancer. However, the precise role of HRGs in gauging the risk and predicting the prognosis of pancreatic adenocarcinoma (PAAD) remains nebulous. METHODS We carried out an elaborate data collection endeavor. Transcriptome data along with pertinent clinical information were obtained from the GSE28735, GSE62452, and TCGA-PAAD cohorts. GWAS data were retrieved from the FinnGen Release 11 and eQTLGen databases. For the drug-target Mendelian randomization (MR) analysis, the "TwoSampleMR" (version 0.5.6) R package was employed. The random survival forest (RSF) model was analyzed using the "randomForestSRC (rfsrc)" R package and further elucidated with the help of the "mlr3" package. Somatic mutation analysis and immune infiltration investigations were conducted by means of the "maftools" (v. 2.12.0) R package and "pRRophetic" R software package, respectively. Targeted drug sensitivity analysis was executed using the "oncopredict" and "parallel" packages. RESULTS Through a meticulous drug-targeted MR analysis and an exhaustive exploration of transcriptome databases (including 2 GSE combat and TCGA cohort), 20 upregulated differentially expressed genes (DEGs) were identified. The RSF model emerged as the optimal choice, and a 9-HRGs signature was selected to construct a prognostic model that boasted an average C-index of 0.777. In the training and validation cohorts, the model exhibited remarkable predictive prowess, with 1-, 2-, and 3-year prediction accuracies of 0.898, 0.932, and 0.922 in the training set, and 0.909, 0.974, and 0.962 in the validation set, respectively. A higher HRG score was found to correlate with adverse events and the N1 stage. Additionally, it was associated with an increase in M0 macrophages and a decline in CD8 + T cell function. For patients with a low HRG score, several commonly used chemotherapeutic agents, namely Gemcitabine, Carboplatin, Sorafenib, and Oxaliplatin, were more efficacious. CONCLUSION The HRG signature holds the potential to serve as effective biomarkers for diagnosing, predicting the prognosis, and assessing the sensitivity to chemotherapy in PAAD.
Collapse
Affiliation(s)
- Chang-Lei Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhi-Yuan Yao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Qu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guan-Ming Shao
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yu-Kun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiang-Yu Pei
- College of Medicine, Qingdao University, Qingdao, China
| | - Jing-Yu Cao
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Zu-Sen Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
12
|
Du H, Zhang L, Feng J, Zhou M, Yang C. Enhancing Photodynamic Therapy Efficacy via Photo-Triggered Calcium Overload and Oxygen Delivery in Tumor Hypoxia Management. ACS APPLIED MATERIALS & INTERFACES 2025; 17:8403-8416. [PMID: 39853325 DOI: 10.1021/acsami.4c21452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Background: Photodynamic therapy (PDT) has emerged as a promising treatment for cancer, primarily due to its ability to generate reactive oxygen species (ROS) that directly induce tumor cell death. However, the hypoxic microenvironment commonly found within tumors poses a significant challenge by inhibiting ROS production. This study aims to investigate the effect of improving tumor hypoxia on enhancing PDT. Result: We employed polylactic-co-glycolic acid (PLGA) as a delivery vector for the encapsulation of indocyanine green (ICG), a photosensitizer, and perfluorohexane (PFH), with surface labeling mannose to facilitate targeted delivery. A potential therapeutic nanoplatform was fabricated, designated as Man-PFH-ICG@PLGA. These nanospheres are capable of localizing at tumor sites and can be tracked using photoacoustic (PA) imaging. Upon laser irradiation, the ROS generated by PDT activated the transient receptor potential cation channel subfamily A member 1 (TRPA1) located on the cell membrane. This activation led to an influx of extracellular Ca2+ and subsequently resulted in calcium overload. The excessive Ca2+ selectively accumulated in mitochondria, disrupting the function of enzymes involved in the mitochondrial respiratory chain. This disruption inhibits cellular respiration and decreases oxygen consumption in tumor cells, ultimately contributing to the alleviation of the hypoxic microenvironment within tumors. Simultaneously, PFH exhibited a high affinity for oxygen and can deliver exogenous oxygen directly to the tumor site through simple diffusion along the concentration gradient. Both the direct and indirect mechanisms synergistically contribute to ameliorating the hypoxic conditions within tumors, thereby augmenting the efficacy of PDT. Conclusions: The synergistic effect of photocontrolled calcium overload from endogenous sources and the oxygen-carrying nanoplatform alleviates tumor hypoxia, thereby enhancing the efficacy of PDT. This approach provides a new perspective on PDT.
Collapse
Affiliation(s)
- Han Du
- Department of Ultrasound, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, China
| | - Liang Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, China
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Jingjing Feng
- Department of Ultrasound, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, China
| | - Mingli Zhou
- Department of Ultrasound, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, China
| | - Chunjiang Yang
- Department of Ultrasound, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
13
|
Mesas C, Moreno J, Doello K, Peña M, López-Romero JM, Prados J, Melguizo C. Cannabidiol effects in stem cells: A systematic review. Biofactors 2025; 51:e2148. [PMID: 39653426 DOI: 10.1002/biof.2148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024]
Abstract
Stem cells play a critical role in human tissue regeneration and repair. In addition, cancer stem cells (CSCs), subpopulations of cancer cells sharing similar characteristics as normal stem cells, are responsible for tumor metastasis and resistance to chemo- and radiotherapy and to tumor relapse. Interestingly, all stem cells have cannabinoid receptors, such as cannabidiol (CBD), that perform biological functions. The aim of this systematic review was to analyze the effect of CBD on both somatic stem cells (SSCs) and CSCs. Of the 276 articles analyzed, 38 were selected according to the inclusion and exclusion criteria. A total of 27 studied the effect of CBD on SSCs, finding that 44% focused on CBD differentiation effect and 56% on its protective activity. On the other hand, 11 articles looked at the effect of CBD on CSCs, including glioblastoma (64%), lung cancer (27%), and breast cancer (only one article). Our results showed that CBD exerted a differentiating and protective effect on SCCs. In addition, this molecule demonstrated an antiproliferative effect on some CSCs, although most of the analyses were performed in vitro. Therefore, although in vivo studies should be necessary to justify its clinical use, CBD and its receptors could be a specific target to act on both SSCs and CSCs.
Collapse
Affiliation(s)
- Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (ibs.GRANADA), Granada, Spain
| | - Javier Moreno
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Kevin Doello
- Instituto de Investigación Biosanitaria de Granada, (ibs.GRANADA), Granada, Spain
- Service of Medical Oncology, Hospital Virgen de las Nieves, Granada, Spain
| | - Mercedes Peña
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (ibs.GRANADA), Granada, Spain
| | - Juan M López-Romero
- Department of Organic Chemistry, Faculty of Sciences, University of Malaga, Málaga, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (ibs.GRANADA), Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| |
Collapse
|
14
|
Yang JL, Yang J, Fang RF, Sai WL, Yao DF, Yao M. Hypoxia upregulates hepatic angiopoietin-2 transcription to promote the progression of hepatocellular carcinoma. World J Hepatol 2024; 16:1480-1492. [DOI: 10.4254/wjh.v16.i12.1480] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Angiopoietin-2 (Ang-2) level is related to hepatocellular carcinoma (HCC) progression. However, the dynamic expression and regulatory mechanism of Ang-2 remain unclear.
AIM To investigate Ang-2 levels in chronic liver diseases and validate early monitoring value with a dynamic model in hepatocarcinogenesis.
METHODS Sprague-Dawley rats in hepatocarcinogenesis were induced with diet 2-fluorenylacet-amide, and grouped based on liver histopathology by hematoxylin and eosin staining. Differently expressed genes or Ang-2 mRNA in livers were analyzed by whole-genome microarray. Ang-2 levels in chronic liver diseases were detected by an enzyme-linked immunosorbent assay.
RESULTS Clinical observation reveled that the circulating levels of Ang-2 and hypoxia-inducible factor-1α (HIF-1α) in patients with chronic liver diseases were progressively increased from benign to HCC (P < 0.001). Dynamic model validated that the up-regulated Ang-2 in liver and blood was positively correlated with HIF-1α in hepatocarcinogenesis (P < 0.001). Mechanistically, Ang-2 was regulated by HIF-1α. When specific HIF-1α- microRNAs transfected into HCC cells, the cell proliferation significantly inhibited, HIF-1α and Ang-2 down-regulated, and also affected epithelial-mesenchymal transition via increasing E-cadherin to block cell invasion or migration with reducing of snail, twist and vimentin.
CONCLUSION Hypoxia-induced Ang-2 up-regulating expression might serve as a sensitive early monitoring biomarker for hepatocarcinogenesis or HCC metastasis.
Collapse
Affiliation(s)
- Jun-Ling Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University and Department of Immunology, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Jie Yang
- Department of Biology, Life Science School of Nantong University, Nantong 226009, Jiangsu Province, China
| | - Rong-Fei Fang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Wen-Li Sai
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University and Department of Immunology, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Deng-Fu Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University and Department of Immunology, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Min Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University and Department of Immunology, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
15
|
Hong J, Du K, Zhang W, Jiang Y, Yu H, Pan T, Wu T, Zhao L, Du W, Zheng SS, Jin H, Chen Y, Cao L. PFOS and Its Commercial Alternative, 6:2 Cl-PFESA, Induce Multidrug Resistance in Pancreatic Cancer. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:22027-22038. [PMID: 39644250 DOI: 10.1021/acs.est.4c08669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS), specifically perfluorooctanesulfonate (PFOS) and its alternative, 2-[(6-chloro-1,1,2,2,3,3,4,4,5,5,6,6-dodecafluorohexyl)oxy]-1,1,2,2-tetrafluoroethanesulfonic acid (6:2 Cl-PFESA), are associated with environmental health concerns and potential cancer progression. However, their impact on multidrug resistance (MDR) in pancreatic cancer (PC) chemotherapy remains unclear. Here, we employed drug-sensitivity assays, including IC50 calculations, in vitro and in vivo models with various chemotherapeutics, and paclitaxel (PTX) as a representative agent, combined with transcriptomic/proteomic sequencing and clinical prognostic analysis, to identify MDR-related genes and validate their relevance, with the objective of establishing the correlation between PFOS/6:2 Cl-PFESA exposure and MDR in PC at molecular, cellular, and animal model levels. Our findings demonstrate that PFOS/6:2 Cl-PFESA exposure increases the drug IC50 in three different PC cell lines for various chemotherapeutic agents. Compared with PFOS, 6:2 Cl-PFESA demonstrated a more pro-MDR effect on PC cells in vitro. In vivo experiments further revealed that PFOS/6:2 Cl-PFESA exposures significantly reduced the efficacy of PTX in PC, with inhibition rates dropping from 78.3% to 23.8%/6.1%, respectively (p < 0.05). This effect was driven by the aberrant activation of the PI3K-ABCB1 pathway, with 6:2 Cl-PFESA demonstrating a stronger capacity to promote this signal pathway's expression and function compared with PFOS. These data suggest that exposure to PFAS may elevate the risk of MDR and subsequent disease progression. Although marketed as a safer alternative to PFOS, the notable impact of 6:2 Cl-PFESA on MDR highlights the necessity for a comprehensive assessment of its potential carcinogenic risks.
Collapse
Affiliation(s)
- Jiawei Hong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, PR China
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, PR China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, PR China
| | - Keyi Du
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, PR China
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, PR China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, PR China
| | - Weichen Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, PR China
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, PR China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, PR China
| | - Yifan Jiang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, PR China
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, PR China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, PR China
| | - Hanxi Yu
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Tingting Pan
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, PR China
| | - Tong Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, PR China
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, PR China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, PR China
| | - Liang Zhao
- Chinese Research Academy of Environmental Sciences, Beijing 100012, PR China
| | - Wei Du
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Shu-Sen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, PR China
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, PR China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, PR China
| | - Hangbiao Jin
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, PR China
- Innovation Research Center of Advanced Environmental Technology, Eco-Industrial Innovation Institute, Zhejiang University of Technology, Quzhou, Zhejiang 324400, PR China
| | - Yuanchen Chen
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, PR China
- Innovation Research Center of Advanced Environmental Technology, Eco-Industrial Innovation Institute, Zhejiang University of Technology, Quzhou, Zhejiang 324400, PR China
| | - Linping Cao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, PR China
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, PR China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, PR China
| |
Collapse
|
16
|
Zhang N, Zhang H, Li S, Wu W, Luo P, Liu Z, Chen Y, Xia Z, Huang C, Cheng Q. Uncovering the predictive and immunomodulatory potential of transient receptor potential melastatin family-related CCNE1 in pan-cancer. Mol Cancer 2024; 23:258. [PMID: 39551726 PMCID: PMC11572178 DOI: 10.1186/s12943-024-02169-7] [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: 06/16/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024] Open
Abstract
Millions of new cases of cancer are diagnosed worldwide each year, making it a serious public health concern. Developments in customized therapy and early detection have significantly enhanced treatment for and results from cancer. Therefore, it is important to investigate new molecular biomarkers. In this study, we created an efficient transient receptor potential melastatin (TRPM) family members-related TRPM-Score for 17 solid tumors. CCNE1, produced from TRPM-Score, was found to be an exceptional biomarker through several sophisticated machine learning and deep learning computational techniques. TRPM-Score and CCNE1 immunotherapeutic prediction, immunological characteristics, and predictive value were thoroughly assessed. In most cancer types, CCNE1 was a substantially dangerous marker. Additional in vitro tests validated CCNE1's immunomodulatory properties, demonstrating that silencing impeded macrophage movement and decreased PD-L1 expression. Additionally, CCNE1 may accurately predict responses to cancer immunotherapy. These findings indicate that the TRPM family-particularly CCNE1, which is associated with TRPM-is a significant player in the pan-cancer domain and can be utilized as a therapeutic target and prognostic biomarkers, especially in immuno-oncology. The thorough characterization of the TRPM family and the discovery of CCNE1 as a crucial downstream effector mark important developments in our comprehension of pan-cancer biology.
Collapse
Affiliation(s)
- Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shuyu Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wantao Wu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Chenshen Huang
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Xiangya Hospital, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.
| |
Collapse
|
17
|
Chen X, Yang Y, Zhou Z, Yu H, Zhang S, Huang S, Wei Z, Ren K, Jin Y. Unraveling the complex interplay between Mitochondria-Associated Membranes (MAMs) and cardiovascular Inflammation: Molecular mechanisms and therapeutic implications. Int Immunopharmacol 2024; 141:112930. [PMID: 39146786 DOI: 10.1016/j.intimp.2024.112930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/26/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
Cardiovascular diseases (CVDs) represent a significant public health concern because of their associations with inflammation, oxidative stress, and abnormal remodeling of the heart and blood vessels. In this review, we discuss the intricate interplay between mitochondria-associated membranes (MAMs) and cardiovascular inflammation, highlighting their role in key cellular processes such as calcium homeostasis, lipid metabolism, oxidative stress management, and ERS. We explored how these functions impact the pathogenesis and progression of various CVDs, including myocardial ischemia-reperfusion injury, atherosclerosis, diabetic cardiomyopathy, cardiovascular aging, heart failure, and pulmonary hypertension. Additionally, we examined current therapeutic strategies targeting MAM-related pathways and proteins, emphasizing the potential of MAMs as therapeutic targets. Our review aims to provide new insights into the mechanisms of cardiovascular inflammation and propose novel therapeutic approaches to improve cardiovascular health outcomes.
Collapse
Affiliation(s)
- Xing Chen
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yang Yang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zheng Zhou
- Department of Geriatric Endocrinology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Haihan Yu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Shuwei Zhang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Siyuan Huang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Ziqing Wei
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
18
|
Bai W, Yan C, Yang Y, Sang L, Hao Q, Yao X, Zhang Y, Yu J, Wang Y, Li X, Meng M, Yang J, Shen J, Sun Y, Sun J. EGF/EGFR-YAP1/TEAD2 signaling upregulates STIM1 in vemurafenib resistant melanoma cells. FEBS J 2024; 291:4969-4983. [PMID: 39298503 DOI: 10.1111/febs.17272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/26/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024]
Abstract
Stromal interaction molecule 1 (STIM1) is the endoplasmic reticulum Ca2+ sensor for store-operated calcium entry and is closely associated with carcinogenesis and tumor progression. Previously, we found that STIM1 is upregulated in melanoma cells resistant to the serine/threonine-protein kinase B-raf inhibitor vemurafenib, although the mechanism underlying this upregulation is unknown. Here, we show that vemurafenib resistance upregulates STIM1 through an epidermal growth factor (EGF)/epidermal growth factor receptor (EGFR)-Yes-associated protein 1 (YAP1)/TEA domain transcription factor 2 (TEAD2) signaling axis. Vemurafenib resistance can lead to an increase in EGF and EGFR levels, causing activation of the EGFR signaling pathway, which promotes YAP1 nuclear localization to increase the expression of STIM1. Our findings not only reveal the mechanism by which vemurafenib resistance promotes STIM1 upregulation, but also provide a rationale for combined targeting of the EGF/EGFR-YAP1/TEAD2-STIM1 axis to improve the therapeutic efficacy of BRAF inhibitor in melanoma patients.
Collapse
Affiliation(s)
- Weiyu Bai
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Chenghao Yan
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Yichen Yang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, China
| | - Lei Sang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Qinggang Hao
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Xinyi Yao
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Yingru Zhang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Jia Yu
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Yifan Wang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Xiaowen Li
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Mingyao Meng
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Jilong Yang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, China
| | - Junling Shen
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Yan Sun
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, China
| | - Jianwei Sun
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| |
Collapse
|
19
|
Leili FR, Shali N, Sheibani M, Jafarian MJ, Pashizeh F, Gerami R, Iraj F, Lashkarshekan AA. Detailed pathological role of non-coding RNAs (ncRNAs) in regulating drug resistance of glioblastoma, and update. Pathol Res Pract 2024; 263:155590. [PMID: 39326365 DOI: 10.1016/j.prp.2024.155590] [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: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024]
Abstract
Glioma is a kind of brain tumor that develops in the central nervous system and is classified based on its histology and molecular genetic features. The lifespan of patients does not exceed 22 months. One of the motives for the low effectiveness of glioma treatment is its radioresistance and chemoresistance. Noncoding RNAs (ncRNAs) are a diverse set of transcripts that do not undergo translation to become proteins in glioma. The ncRNAs have been identified as significant regulators of several biological processes in different cell types and tissues, and their abnormal function has been linked to glioma. They are known to impact important occurrences, including carcinogenesis, progression, and enhanced treatment resistance in glioma cells. The ncRNAs control cell proliferation, migration, epithelial-to-mesenchymal transition (EMT), invasion, and drug resistance in glioma cells. The main focus of this study is to inspect the involvement of ncRNAs in the drug resistance of glioma.
Collapse
Affiliation(s)
- Foad Rahmanpour Leili
- Department of Neurology Faculty of Medicine, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Niloofar Shali
- Department of Clinical Biochemistry, School of Medicine, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | - Mehrnaz Sheibani
- Division of Pediatric Neurology, University of Tabriz, Tabriz, Iran
| | | | - Fatemeh Pashizeh
- Department of Immunology, School of Medicine, Shahid Sadoughi University of Medical Science, Yazd 8916188635, Iran
| | - Reza Gerami
- Department of Radiology, Faculty of Medicine, AJA University of Medical Science, Tehran, Iran.
| | | | | |
Collapse
|
20
|
Koskimäki S, Tojkander S. TRPV4-A Multifunctional Cellular Sensor Protein with Therapeutic Potential. SENSORS (BASEL, SWITZERLAND) 2024; 24:6923. [PMID: 39517820 PMCID: PMC11548305 DOI: 10.3390/s24216923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/02/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
Transient receptor potential vanilloid (TRPV) channel proteins belong to the superfamily of TRP proteins that form cationic channels in the animal cell membranes. These proteins have various subtype-specific functions, serving, for example, as sensors for pain, pressure, pH, and mechanical extracellular stimuli. The sensing of extracellular cues by TRPV4 triggers Ca2+-influx through the channel, subsequently coordinating numerous intracellular signaling cascades in a spatio-temporal manner. As TRPV channels play such a wide role in various cellular and physiological functions, loss or impaired TRPV protein activity naturally contributes to many pathophysiological processes. This review concentrates on the known functions of TRPV4 sensor proteins and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Sanna Koskimäki
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | | |
Collapse
|
21
|
Wilczyński B, Dąbrowska A, Kulbacka J, Baczyńska D. Chemoresistance and the tumor microenvironment: the critical role of cell-cell communication. Cell Commun Signal 2024; 22:486. [PMID: 39390572 PMCID: PMC11468187 DOI: 10.1186/s12964-024-01857-7] [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: 07/08/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
Resistance of cancer cells to anticancer drugs remains a major challenge in modern medicine. Understanding the mechanisms behind the development of chemoresistance is key to developing appropriate therapies to counteract it. Nowadays, with advances in technology, we are paying more and more attention to the role of the tumor microenvironment (TME) and intercellular interactions in this process. We also know that important elements of the TME are not only the tumor cells themselves but also other cell types, such as mesenchymal stem cells, cancer-associated fibroblasts, stromal cells, and macrophages. TME elements can communicate with each other indirectly (via cytokines, chemokines, growth factors, and extracellular vesicles [EVs]) and directly (via gap junctions, ligand-receptor pairs, cell adhesion, and tunnel nanotubes). This communication appears to be critical for the development of chemoresistance. EVs seem to be particularly interesting structures in this regard. Within these structures, lipids, proteins, and nucleic acids can be transported, acting as signaling molecules that interact with numerous biochemical pathways, thereby contributing to chemoresistance. Moreover, drug efflux pumps, which are responsible for removing drugs from cancer cells, can also be transported via EVs.
Collapse
Affiliation(s)
- Bartosz Wilczyński
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, Wroclaw, 50-367, Poland
| | - Alicja Dąbrowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, Wroclaw, 50-367, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, Wroclaw, 50-556, Poland.
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, Vilnius, LT-08406, Lithuania.
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, Wroclaw, 50-556, Poland
| |
Collapse
|
22
|
Qi H, Ma X, Ma Y, Jia L, Liu K, Wang H. Mechanisms of HIF1A-mediated immune evasion in gastric cancer and the impact on therapy resistance. Cell Biol Toxicol 2024; 40:87. [PMID: 39384651 PMCID: PMC11464584 DOI: 10.1007/s10565-024-09917-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 09/04/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND The high prevalence and detrimental effects on patient outcomes make gastric cancer (GC) a significant health issue that persists internationally. Existing treatment modalities exhibit limited efficacy, prompting the exploration of immune checkpoint inhibitors as a novel therapeutic approach. However, resistance to immunotherapy poses a significant challenge in GC management, necessitating a profound grasp of the intrinsic molecular pathways. METHODS This study focuses on investigating the immunosuppressive mechanisms of quiescent cancer cells (QCCs) in GC, particularly their resistance to T-cell-mediated immune responses. Utilizing mouse models, gene editing techniques, and transcriptome sequencing, we aim to elucidate the interactions between QCCs, immune cells, and key regulatory factors like HIF1A. Functional enrichment analysis will further underscore the role of glycolysis-related genes in mediating immunosuppression by QCCs. RESULTS The cancer cells that survived GC treated with T-cell therapy lost their proliferative ability. QCCs, as the main resistance force to immunotherapy, exhibit stronger resistance to CD8+ T-cell attack and possess higher cancer-initiating potential. Single-cell sequencing analysis revealed that the microenvironment in the QCCs region harbors more M2-type tumor-associated macrophages and fewer T cells. This microenvironment in the QCCs region leads to the downregulation of T-cell immune activation and alters macrophage metabolic function. Transcriptome sequencing of QCCs identified upregulated genes related to chemo-resistance, hypoxia, and glycolysis. In vitro cell experiments illustrated that HIF1A promotes the transcription of glycolysis-related genes, and silencing HIF1A in QCCs enhances T-cell proliferation and activation in co-culture systems, induces apoptosis in QCCs, and increases QCCs' sensitivity to immune checkpoint inhibitors. In vivo, animal experiments showed that silencing HIF1A in QCCs can inhibit GC growth and metastasis. CONCLUSION Unraveling the molecular mechanisms by which QCCs resist T-cell-mediated immune responses through immunosuppression holds promising implications for refining treatment strategies and enhancing patient outcomes in GC. By delineating these intricate interactions, this study contributes crucial insights into precision medicine and improved therapeutic outcomes in GC management.
Collapse
Affiliation(s)
- Hao Qi
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Xiaoyu Ma
- Departments of Gastrointestinal Endoscopy, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yu Ma
- Department of Nuclear Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Liuyu Jia
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Kuncong Liu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Honghu Wang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
23
|
Wang Y, Su L, Hu Z, Peng S, Li N, Fu H, Wang B, Wu H. Resveratrol suppresses liver cancer progression by downregulating AKR1C3: targeting HCC with HSA nanomaterial as a carrier to enhance therapeutic efficacy. Apoptosis 2024; 29:1429-1453. [PMID: 39023830 DOI: 10.1007/s10495-024-01995-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
The enzyme AKR1C3 plays a crucial role in hormone and drug metabolism and is associated with abnormal expression in liver cancer, leading to tumor progression and poor prognosis. Nanoparticles modified with HSA can modulate the tumor microenvironment by enhancing photodynamic therapy to induce apoptosis in tumor cells and alleviate hypoxia. Therefore, exploring the potential regulatory mechanisms of resveratrol on AKR1C3 through the construction of HSA-RSV NPs carriers holds significant theoretical and clinical implications for the treatment of liver cancer. The aim of this study is to investigate the targeted regulation of AKR1C3 expression through the loading of resveratrol (RSV) on nanomaterials HSA-RSV NPs (Nanoparticles) in order to alleviate tumor hypoxia and inhibit the progression of hepatocellular carcinoma (HCC), and to explore its molecular mechanism. PubChem database and PharmMapper server were used to screen the target genes of RSV. HCC-related differentially expressed genes (DEGs) were analyzed through the GEO dataset, and relevant genes were retrieved from the GeneCards database, resulting in the intersection of the three to obtain candidate DEGs. GO and KEGG enrichment analyses were performed on the candidate DEGs to analyze the potential cellular functions and molecular signaling pathways affected by the main target genes. The cytohubba plugin was used to screen the top 10 target genes ranked by Degree and further intersected the results of LASSO and Random Forest (RF) to obtain hub genes. The expression analysis of hub genes and the prediction of malignant tumor prognosis were conducted. Furthermore, a pharmacophore model was constructed using PharmMapper. Molecular docking simulations were performed using AutoDockTools 1.5.6 software, and ROC curve analysis was performed to determine the core target. In vitro cell experiments were carried out by selecting appropriate HCC cell lines, treating HCC cells with different concentrations of RSV, or silencing or overexpressing AKR1C3 using lentivirus. CCK-8, clone formation, flow cytometry, scratch experiment, and Transwell were used to measure cancer cell viability, proliferation, migration, invasion, and apoptosis, respectively. Cellular oxygen consumption rate was analyzed using the Seahorse XF24 analyzer. HSA-RSV NPs were prepared, and their characterization and cytotoxicity were evaluated. The biological functional changes of HCC cells after treatment were detected. An HCC subcutaneous xenograft model was established in mice using HepG2 cell lines. HSA-RSV NPs were injected via the tail vein, with a control group set, to observe changes in tumor growth, tumor targeting of NPs, and biological safety. TUNEL, Ki67, and APC-hypoxia probe staining were performed on excised tumor tissue to detect tumor cell proliferation, apoptosis, and hypoxia. Lentivirus was used to silence or overexpress AKR1C3 simultaneously with the injection of HSA-RSV NPs via the tail vein to assess the impact of AKR1C3 on the regulation of HSA-RSV NPs in HCC progression. Bioinformatics analysis revealed that AKR1C3 is an important target gene involved in the regulation of HCC by RSV, which is associated with the prognosis of HCC patients and upregulated in expression. In vitro cell experiments showed that RSV significantly inhibits the respiratory metabolism of HCC cells, suppressing their proliferation, migration, and invasion and promoting apoptosis. Silencing AKR1C3 further enhances the toxicity of RSV towards HCC cells. The characterization and cytotoxicity experiments of nanomaterials demonstrated the successful construction of HSA-RSV NPs, which exhibited stronger inhibitory effects on HCC cells. In vivo, animal experiments further confirmed that targeted downregulation of AKR1C3 by HSA-RSV NPs suppresses the progression of HCC and tumor hypoxia while exhibiting tumor targeting and biological safety. Targeted downregulation of AKR1C3 by HSA-RSV NPs can alleviate HCC tumor hypoxia and inhibit the progression of HCC.
Collapse
Affiliation(s)
- Ying Wang
- Operating Room, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Longxiang Su
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Zhansheng Hu
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Shuang Peng
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Na Li
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Haiyan Fu
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Baoquan Wang
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China
| | - Huiping Wu
- Intensive Care Unit, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121001, China.
| |
Collapse
|
24
|
Lv W, Wang Y. Neural Influences on Tumor Progression Within the Central Nervous System. CNS Neurosci Ther 2024; 30:e70097. [PMID: 39469896 PMCID: PMC11519750 DOI: 10.1111/cns.70097] [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: 07/08/2024] [Revised: 09/21/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
For decades, researchers have studied how brain tumors, the immune system, and drugs interact. With the advances in cancer neuroscience, which centers on defining and therapeutically targeting nervous system-cancer interactions, both within the local tumor microenvironment (TME) and on a systemic level, the subtle relationship between neurons and tumors in the central nervous system (CNS) has been deeply studied. Neurons, as the executors of brain functional activities, have been shown to significantly influence the emergence and development of brain tumors, including both primary and metastatic tumors. They engage with tumor cells via chemical or electrical synapses, directly regulating tumors or via intricate coupling networks, and also contribute to the TME through paracrine signaling, secreting proteins that exert regulatory effects. For instance, in a study involving a mouse model of glioblastoma, the authors observed a 42% increase in tumor volume when neuronal activity was stimulated, compared to controls (p < 0.01), indicating a direct correlation between neural activity and tumor growth. These thought-provoking results offer promising new strategies for brain tumor therapies, highlighting the potential of neuronal modulation to curb tumor progression. Future strategies may focus on developing drugs to inhibit or neutralize proteins and other bioactive substances secreted by neurons, break synaptic connections and interactions between infiltrating cells and tumor cells, as well as disrupt electrical coupling within glioma cell networks. By harnessing the insights gained from this research, we aspire to usher in a new era of brain tumor therapies that are both more potent and precise.
Collapse
Affiliation(s)
- Wenhao Lv
- Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouZhejiangChina
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
25
|
Liu ZQ, Zhang Q, Liu YL, Yu XQ, Chui RH, Zhang LL, Zhao B, Ma LY. Recent contributions of pyridazine as a privileged scaffold of anticancer agents in medicinal chemistry: An updated review. Bioorg Med Chem 2024; 111:117847. [PMID: 39121679 DOI: 10.1016/j.bmc.2024.117847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
Pyridazine, as a privileged scaffold, has been extensively utilized in drug development due to its multiple biological activities. Especially around its distinctive anticancer property, a massive number of pyridazine-containing compounds have been synthesized and evaluated that target a diverse array of biological processes involved in cancer onset and progression. These include glutaminase 1 (GLS1) inhibitors, tropomyosin receptor kinase (TRK) inhibitors, and bromodomain containing protein (BRD) inhibitors, targeting aberrant tumor metabolism, cell signal transduction and epigenetic modifications, respectively. Pyridazine moieties functioned as either core frameworks or warheads in the above agents, exhibiting promising potential in cancer treatment. Therefore, the review aims to summarize the recent contributions of pyridazine derivatives as potent anticancer agents between 2020 and 2024, focusing mainly on their structure-activity relationships (SARs) and development strategies, with a view to show that the application of the pyridazine scaffold by different medicinal chemists provides new insights into the rational design of anticancer drugs.
Collapse
Affiliation(s)
- Zi-Qiang Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Qian Zhang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Yu-Lin Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Xiao-Qian Yu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Rui-Hao Chui
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Lin-Lin Zhang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Bing Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Li-Ying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China; China Meheco Topfond Pharmaceutical Co., Key Laboratory of Cardio-cerebrovascular Drug, Zhumadian 463000, PR China.
| |
Collapse
|
26
|
Yang D. TRPA1-Related Diseases and Applications of Nanotherapy. Int J Mol Sci 2024; 25:9234. [PMID: 39273183 PMCID: PMC11395144 DOI: 10.3390/ijms25179234] [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: 07/07/2024] [Revised: 07/30/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Transient receptor potential (TRP) channels, first identified in Drosophila in 1969, are multifunctional ion channels expressed in various cell types. Structurally, TRP channels consist of six membrane segments and are classified into seven subfamilies. Transient receptor potential ankyrin 1 (TRPA1), the first member of the TRPA family, is a calcium ion affinity non-selective cation channel involved in sensory transduction and responds to odors, tastes, and chemicals. It also regulates temperature and responses to stimuli. Recent studies have linked TRPA1 to several disorders, including chronic pain, inflammatory diseases, allergies, and respiratory problems, owing to its activation by environmental toxins. Mutations in TRPA1 can affect the sensory nerves and microvasculature, potentially causing nerve pain and vascular problems. Understanding the function of TRPA1 is important for the development of treatments for these diseases. Recent developments in nanomedicines that target various ion channels, including TRPA1, have had a significant impact on disease treatment, providing innovative alternatives to traditional disease treatments by overcoming various adverse effects.
Collapse
Affiliation(s)
- Dongki Yang
- Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
27
|
Wang Y, Li N, Qu L, Zhang M, Li Z, Li X, Cai D. Hemoglobin nanoclusters-mediated regulation of KPNA4 in hypoxic tumor microenvironment enhances photodynamic therapy in hepatocellular carcinoma. J Nanobiotechnology 2024; 22:473. [PMID: 39135024 PMCID: PMC11318167 DOI: 10.1186/s12951-024-02717-9] [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: 05/10/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a highly malignant tumor known for its hypoxic environment, which contributes to resistance against the anticancer drug Sorafenib (SF). Addressing SF resistance in HCC requires innovative strategies to improve tumor oxygenation and effectively deliver therapeutics. RESULTS In our study, we explored the role of KPNA4 in mediating hypoxia-induced SF resistance in HCC. We developed hemoglobin nanoclusters (Hb-NCs) capable of carrying oxygen, loaded with indocyanine green (ICG) and SF, named HPRG@SF. In vitro, HPRG@SF targeted HCC cells, alleviated hypoxia, suppressed KPNA4 expression, and enhanced the cytotoxicity of PDT against hypoxic, SF-resistant HCC cells. In vivo experiments supported these findings, showing that HPRG@SF effectively improved the oxygenation within the tumor microenvironment and countered SF resistance through combined photodynamic therapy (PDT). CONCLUSION The combination of Hb-NCs with ICG and SF, forming HPRG@SF, presents a potent strategy to overcome drug resistance in hepatocellular carcinoma by improving hypoxia and employing PDT. This approach not only targets the hypoxic conditions that underlie resistance but also provides a synergistic anticancer effect, highlighting its potential for clinical applications in treating resistant HCC.
Collapse
Affiliation(s)
- Yiliang Wang
- Department of Anesthesiology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Nu Li
- Department of breast surgery, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Letian Qu
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Mu Zhang
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Zhuo Li
- The Fourth People's Hospital of Shenyang, 110002, Liaoning Province, China
| | - Xiang Li
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.
| | - Dasheng Cai
- Department of Anesthesiology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
28
|
Płonka-Czerw J, Żyrek L, Latocha M. Changes in the Sensitivity of MCF-7 and MCF-7/DX Breast Cancer Cells to Cytostatic in the Presence of Metformin. Molecules 2024; 29:3531. [PMID: 39124936 PMCID: PMC11313889 DOI: 10.3390/molecules29153531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Multidrug resistance is a serious problem in modern medicine and the reason for the failure of various therapies. A particularly important problem is the occurrence of multidrug resistance in cancer therapies which affects many cancer patients. Observations on the effect of metformin-a well-known hypoglycemic drug used in the treatment of type 2 diabetes-on cancer cells indicate the possibility of an interaction of this substance with drugs already used and, as a result, an increase in the sensitivity of cancer cells to cytostatics. The aim of this study was to evaluate the effect of metformin on the occurrence of multidrug resistance of breast cancer cells. The MCF-7-sensitive cell line and the MCF-7/DX cytostatic-resistant cell line were used for this study. WST-1 and LDH assays were used to evaluate the effects of metformin and doxorubicin on cell proliferation and viability. The effect of metformin on increasing the sensitivity of MCF-7 and MCF-7/DX cells to doxorubicin was evaluated in an MDR test. The participation of metformin in increasing the sensitivity of resistant cells to the effect of the cytostatic (doxorubicin) has been demonstrated.
Collapse
Affiliation(s)
- Justyna Płonka-Czerw
- Department of Cell Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland; (L.Ż.); (M.L.)
| | | | | |
Collapse
|
29
|
Jiang Z, Fang Z, Hong D, Wang X. Cancer Immunotherapy with "Vascular-Immune" Crosstalk as Entry Point: Associated Mechanisms, Therapeutic Drugs and Nano-Delivery Systems. Int J Nanomedicine 2024; 19:7383-7398. [PMID: 39050878 PMCID: PMC11268745 DOI: 10.2147/ijn.s467222] [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: 03/05/2024] [Accepted: 07/04/2024] [Indexed: 07/27/2024] Open
Abstract
Tumor vessels characterized by abnormal functions and structures hinder the infiltration and immune antigen presentation of immune cells by inducing the formation of an immunosuppressive microenvironment ("cold" environment). Vascular-targeted therapy has been proven to enhance immune stimulation and the effectiveness of immunotherapy by modulating the "cold" microenvironment, such as hypoxia and an acidic microenvironment. Notably, a therapeutic strategy based on "vascular-immune" crosstalk can achieve dual regulation of tumor vessels and the immune system by reprogramming the tumor microenvironment (TME), thus forming a positive feedback loop between tumor vessels and the immune microenvironment. From this perspective, we discuss the factors of tumor angiogenesis and "cold" TME formation. Building on this foundation, some vascular-targeted therapeutic drugs will be elaborated upon in detail to achieve dual regulation of tumor vessels and immunity. More importantly, we focus on cutting-edge nanotechnology in view of "vascular-immune" crosstalk and discuss the rational fabrication of tailor-made nanosystems for efficiently enhancing immunotherapy.
Collapse
Affiliation(s)
- Zhijie Jiang
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, People’s Republic of China
| | - Zhujun Fang
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, People’s Republic of China
| | - Dongsheng Hong
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, People’s Republic of China
| | - Xiaojuan Wang
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, People’s Republic of China
| |
Collapse
|
30
|
Wang F, Fu X, Chang M, Wei T, Lin R, Tong H, Zhang X, Yuan R, Zhou Z, Huang X, Zhang W, Su W, Lu Y, Liang Z, Zhang J. The Interaction of Calcium-Sensing Receptor with KIF11 Enhances Cisplatin Resistance in Lung Adenocarcinoma via BRCA1/cyclin B1 pathway. Int J Biol Sci 2024; 20:3892-3910. [PMID: 39113697 PMCID: PMC11302892 DOI: 10.7150/ijbs.92046] [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: 11/09/2023] [Accepted: 07/05/2024] [Indexed: 08/10/2024] Open
Abstract
Cisplatin (DDP) is commonly used in the treatment of non-small cell lung cancer (NSCLC), including lung adenocarcinoma (LUAD), and the primary cause for its clinical inefficacy is chemoresistance. Here, we aimed to investigate a novel mechanism of chemoresistance in LUAD cells, focusing on the calcium-sensing receptor (CaSR). In this study, high CaSR expression was detected in DDP-resistant LUAD cells, and elevated CaSR expression is strongly correlated with poor prognosis in LUAD patients receiving chemotherapy. LUAD cells with high CaSR expression exhibited decreased sensitivity to cisplatin, and the growth of DDP-resistant LUAD cells was inhibited by cisplatin treatment in combination with CaSR suppression, accompanied by changes in BRCA1 and cyclin B1 protein expression both in vitro and in vivo. Additionally, an interaction between CaSR and KIF11 was identified. Importantly, suppressing KIF11 resulted in decreased protein levels of BRCA1 and cyclin B1, enhancing the sensitivity of DDP-resistant LUAD cells to cisplatin with no obvious decrease in CaSR. Here, our findings established the critical role of CaSR in promoting cisplatin resistance in LUAD cells by modulating cyclin B1 and BRCA1 and identified KIF11 as a mediator, highlighting the potential therapeutic value of targeting CaSR to overcome chemoresistance in LUAD.
Collapse
Affiliation(s)
- Fuhao Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xing Fu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Ming Chang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Tianzi Wei
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Risheng Lin
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Haibo Tong
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Xiao Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Runzhu Yuan
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhiqing Zhou
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xin Huang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Wei Zhang
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Wenmei Su
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yi Lu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zhen Liang
- The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jian Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
31
|
Olszewska AM, Zmijewski MA. Genomic and non-genomic action of vitamin D on ion channels - Targeting mitochondria. Mitochondrion 2024; 77:101891. [PMID: 38692383 DOI: 10.1016/j.mito.2024.101891] [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: 03/12/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/03/2024]
Abstract
Recent studies revealed that mitochondria are not only a place of vitamin D3 metabolism but also direct or indirect targets of its activities. This review summarizes current knowledge on the regulation of ion channels from plasma and mitochondrial membranes by the active form of vitamin D3 (1,25(OH)2D3). 1,25(OH)2D3, is a naturally occurring hormone with pleiotropic activities; implicated in the modulation of cell differentiation, and proliferation and in the prevention of various diseases, including cancer. Many experimental data indicate that 1,25(OH)2D3 deficiency induces ionic remodeling and 1,25(OH)2D3 regulates the activity of multiple ion channels. There are two main theories on how 1,25(OH)2D3 can modify the function of ion channels. First, describes the involvement of genomic pathways of response to 1,25(OH)2D3 in the regulation of the expression of the genes encoding channels, their auxiliary subunits, or additional regulators. Interestingly, intracellular ion channels, like mitochondrial, are encoded by the same genes as plasma membrane channels. Therefore, the comprehensive genomic regulation of the channels from these two different cellular compartments we analyzed using a bioinformatic approach. The second theory explores non-genomic pathways of vitamin D3 activities. It was shown, that 1,25(OH)2D3 indirectly regulates enzymes that impact ion channels, change membrane physical properties, or directly bind to channel proteins. In this article, the involvement of genomic and non-genomic pathways regulated by 1,25(OH)2D3 in the modulation of the levels and activity of plasma membrane and mitochondrial ion channels was investigated by an extensive review of the literature and analysis of the transcriptomic data using bioinformatics.
Collapse
Affiliation(s)
- A M Olszewska
- Department of Histology, Medical University of Gdansk, 1a Debinki, 80-211 Gdansk, Poland
| | - M A Zmijewski
- Department of Histology, Medical University of Gdansk, 1a Debinki, 80-211 Gdansk, Poland.
| |
Collapse
|
32
|
Cheng G, Zhao Y, Sun F, Zhang Q. Novel insights into STIM1's role in store-operated calcium entry and its implications for T-cell mediated inflammation in trigeminal neuralgia. Front Mol Neurosci 2024; 17:1391189. [PMID: 38962804 PMCID: PMC11221526 DOI: 10.3389/fnmol.2024.1391189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 04/30/2024] [Indexed: 07/05/2024] Open
Abstract
This investigation aims to elucidate the novel role of Stromal Interaction Molecule 1 (STIM1) in modulating store-operated calcium entry (SOCE) and its subsequent impact on inflammatory cytokine release in T lymphocytes, thereby advancing our understanding of trigeminal neuralgia (TN) pathogenesis. Employing the Gene Expression Omnibus (GEO) database, we extracted microarray data pertinent to TN to identify differentially expressed genes (DEGs). A subsequent comparison with SOCE-related genes from the Genecards database helped pinpoint potential target genes. The STRING database facilitated protein-protein interaction (PPI) analysis to spotlight STIM1 as a gene of interest in TN. Through histological staining, transmission electron microscopy (TEM), and behavioral assessments, we probed STIM1's pathological effects on TN in rat models. Additionally, we examined STIM1's influence on the SOCE pathway in trigeminal ganglion cells using techniques like calcium content measurement, patch clamp electrophysiology, and STIM1- ORAI1 co-localization studies. Changes in the expression of inflammatory markers (TNF-α, IL-1β, IL-6) in T cells were quantified using Western blot (WB) and enzyme-linked immunosorbent assay (ELISA) in vitro, while immunohistochemistry and flow cytometry were applied in vivo to assess these cytokines and T cell count alterations. Our bioinformatic approach highlighted STIM1's significant overexpression in TN patients, underscoring its pivotal role in TN's etiology and progression. Experimental findings from both in vitro and in vivo studies corroborated STIM1's regulatory influence on the SOCE pathway. Furthermore, STIM1 was shown to mediate SOCE-induced inflammatory cytokine release in T lymphocytes, a critical factor in TN development. Supportive evidence from histological, ultrastructural, and behavioral analyses reinforced the link between STIM1-mediated SOCE and T lymphocyte-driven inflammation in TN pathogenesis. This study presents novel evidence that STIM1 is a key regulator of SOCE and inflammatory cytokine release in T lymphocytes, contributing significantly to the pathogenesis of trigeminal neuralgia. Our findings not only deepen the understanding of TN's molecular underpinnings but also potentially open new avenues for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Guangyu Cheng
- Translational Medicine Research Center of Traditional Chinese Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu Zhao
- Department of Acupuncture, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fujia Sun
- Department of Acupuncture, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qi Zhang
- Preventive Treatment Center, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
33
|
Kohashi H, Nagata R, Tamenori Y, Amatani T, Ueda Y, Mori Y, Kasahara Y, Obika S, Shimojo M. A novel transient receptor potential C3/C6 selective activator induces the cellular uptake of antisense oligonucleotides. Nucleic Acids Res 2024; 52:4784-4798. [PMID: 38621757 PMCID: PMC11109983 DOI: 10.1093/nar/gkae245] [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: 11/22/2023] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/17/2024] Open
Abstract
Antisense oligonucleotide (ASO) therapy is a novel therapeutic approach in which ASO specifically binds target mRNA, resulting in mRNA degradation; however, cellular uptake of ASOs remains critically low, warranting improvement. Transient receptor potential canonical (TRPC) channels regulate Ca2+ influx and are activated upon stimulation by phospholipase C-generated diacylglycerol. Herein, we report that a novel TRPC3/C6/C7 activator, L687, can induce cellular ASO uptake. L687-induced ASO uptake was enhanced in a dose- and incubation-time-dependent manner. L687 enhanced the knockdown activity of various ASOs both in vitro and in vivo. Notably, suppression of TRPC3/C6 by specific siRNAs reduced ASO uptake in A549 cells. Application of BAPTA-AM, a Ca2+ chelator, and SKF96365, a TRPC3/C6 inhibitor, suppressed Ca2+ influx via TRPC3/C6, resulting in reduced ASO uptake, thereby suggesting that Ca2+ influx via TRPC3/C6 is critical for L687-mediated increased ASO uptake. L687 also induced dextran uptake, indicating that L687 increased endocytosis. Adding ASO to L687 resulted in endosome accumulation; however, the endosomal membrane disruptor UNC7938 facilitated endosomal escape and enhanced knockdown activity. We discovered a new function for TRPC activators regarding ASO trafficking in target cells. Our findings provide an opportunity to formulate an innovative drug delivery system for the therapeutic development of ASO.
Collapse
Affiliation(s)
- Hiroto Kohashi
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Ryu Nagata
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Yusuke Tamenori
- School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Tomorrow Amatani
- Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Yoshifumi Ueda
- Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Yasuo Mori
- Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Yuuya Kasahara
- National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Satoshi Obika
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
- Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka 565-0871, Japan
| | - Masahito Shimojo
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
34
|
Pan T, Gao Y, Xu G, Yu L, Xu Q, Yu J, Liu M, Zhang C, Ma Y, Li Y. Widespread transcriptomic alterations of transient receptor potential channel genes in cancer. Brief Funct Genomics 2024; 23:214-227. [PMID: 37288496 DOI: 10.1093/bfgp/elad023] [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: 02/09/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/09/2023] Open
Abstract
Ion channels, in particular transient-receptor potential (TRP) channels, are essential genes that play important roles in many physiological processes. Emerging evidence has demonstrated that TRP genes are involved in a number of diseases, including various cancer types. However, we still lack knowledge about the expression alterations landscape of TRP genes across cancer types. In this review, we comprehensively reviewed and summarised the transcriptomes from more than 10 000 samples in 33 cancer types. We found that TRP genes were widespreadly transcriptomic dysregulated in cancer, which was associated with clinical survival of cancer patients. Perturbations of TRP genes were associated with a number of cancer pathways across cancer types. Moreover, we reviewed the functions of TRP family gene alterations in a number of diseases reported in recent studies. Taken together, our study comprehensively reviewed TRP genes with extensive transcriptomic alterations and their functions will directly contribute to cancer therapy and precision medicine.
Collapse
Affiliation(s)
- Tao Pan
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Yueying Gao
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Gang Xu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | | | - Qi Xu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Jinyang Yu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Meng Liu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Can Zhang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Yongsheng Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| |
Collapse
|
35
|
Li L, Song Q, Zhou J, Ji Q. Controllers of histone methylation-modifying enzymes in gastrointestinal cancers. Biomed Pharmacother 2024; 174:116488. [PMID: 38520871 DOI: 10.1016/j.biopha.2024.116488] [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: 11/23/2023] [Revised: 02/26/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024] Open
Abstract
Gastrointestinal (GI) cancers have been considered primarily genetic malignancies, caused by a series of progressive genetic alterations. Accumulating evidence shows that histone methylation, an epigenetic modification program, plays an essential role in the different pathological stages of GI cancer progression, such as precancerous lesions, tumorigenesis, and tumor metastasis. Histone methylation-modifying enzymes, including histone methyltransferases (HMTs) and demethylases (HDMs), are the main executor of post-transcriptional modification. The abnormal expression of histone methylation-modifying enzymes characterizes GI cancers with complex pathogenesis and progression. Interactions between upstream controllers and histone methylation-modifying enzymes have recently been revealed, and have provided numerous opportunities to elucidate the pathogenesis of GI cancers in depth and clearly. Here we focus on the association between histone methylation-modifying enzymes and their controllers, aiming to provide a new perspective on the molecular research and clinical management of GI cancers.
Collapse
Affiliation(s)
- Ling Li
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qing Song
- Department of Medical Oncology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215007, China
| | - Jing Zhou
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China.
| | - Qing Ji
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
36
|
Dave B, Patel M, Suresh S, Ginjupalli M, Surya A, Albdour M, Kooner KS. Wound Modulations in Glaucoma Surgery: A Systematic Review. Bioengineering (Basel) 2024; 11:446. [PMID: 38790314 PMCID: PMC11117829 DOI: 10.3390/bioengineering11050446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Excessive fibrosis and resultant poor control of intraocular pressure (IOP) reduce the efficacy of glaucoma surgeries. Historically, corticosteroids and anti-fibrotic agents, such as mitomycin C (MMC) and 5-fluorouracil (5-FU), have been used to mitigate post-surgical fibrosis, but these have unpredictable outcomes. Therefore, there is a need to develop novel treatments which provide increased effectiveness and specificity. This review aims to provide insight into the pathophysiology behind wound healing in glaucoma surgery, as well as the current and promising future wound healing agents that are less toxic and may provide better IOP control.
Collapse
Affiliation(s)
- Bhoomi Dave
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.D.); (M.P.); (S.S.); (M.G.); (A.S.)
- Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Monica Patel
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.D.); (M.P.); (S.S.); (M.G.); (A.S.)
| | - Sruthi Suresh
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.D.); (M.P.); (S.S.); (M.G.); (A.S.)
| | - Mahija Ginjupalli
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.D.); (M.P.); (S.S.); (M.G.); (A.S.)
| | - Arvind Surya
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.D.); (M.P.); (S.S.); (M.G.); (A.S.)
| | - Mohannad Albdour
- Department of Ophthalmology, King Hussein Medical Center Royal Medical Services, Amman 11180, Jordan;
| | - Karanjit S. Kooner
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (B.D.); (M.P.); (S.S.); (M.G.); (A.S.)
- Department of Ophthalmology, Veteran Affairs North Texas Health Care System Medical Center, Dallas, TX 75216, USA
| |
Collapse
|
37
|
Li B, Liu Y, Chen D, Sun S. Comprehensive Analysis of Predictive Value and the potential therapeutic target of NLRP3 inflammasome in glioma based on tumor microenvironment. Clin Immunol 2024; 261:109918. [PMID: 38307475 DOI: 10.1016/j.clim.2024.109918] [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: 11/16/2023] [Revised: 01/09/2024] [Accepted: 01/22/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND Glioma exhibits high recurrence rates and poor prognosis. The nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome plays a crucial role in inflammation. There is a lack of research exploring the NLRP3 in glioma. METHODS We used several databases, networks, Western blotting, multiple immunofluorescence staining to analyze the role of NLRP3 in inflammatory tumor microenvironment (TME). RESULTS NLRP3 is higher-expression in glioma with a low mutation load. NLRP3 expression is linked to the infiltration of immune cells, chemokines, immunomodulators, and the TME. Signaling pathways, co-expression genes and interacting proteins contribute to the up-regulation of NLRP3. Patients responding to immunotherapy positively tend to have lower NLRP3 expression relating to the overall survival based on nomogram. Sensitivity to molecular medicines is observed in relation to NLRP3. CONCLUSION The NLRP3 inflammasome plays a pivotal role in TME which could serve as a higher predictive value biomarker and therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Bihan Li
- Nanjing municipal center for disease control and prevention, Nanjing, Jiangsu, China; Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Ying Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Dawei Chen
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Shilong Sun
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
38
|
Wang Y, Wang B, Cao W, Xu X. TGF-β-activated circRYK drives glioblastoma progression by increasing VLDLR mRNA expression and stability in a ceRNA- and RBP-dependent manner. J Exp Clin Cancer Res 2024; 43:73. [PMID: 38454465 PMCID: PMC10921701 DOI: 10.1186/s13046-024-03000-3] [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: 11/05/2023] [Accepted: 03/01/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND The TGF-β signalling pathway is intricately associated with the progression of glioblastoma (GBM). The objective of this study was to examine the role of circRNAs in the TGF-β signalling pathway. METHODS In our research, we used transcriptome analysis to search for circRNAs that were activated by TGF-β. After confirming the expression pattern of the selected circRYK, we carried out in vitro and in vivo cell function assays. The underlying mechanisms were analysed via RNA pull-down, luciferase reporter, and RNA immunoprecipitation assays. RESULTS CircRYK expression was markedly elevated in GBM, and this phenotype was strongly associated with a poor prognosis. Functionally, circRYK promotes epithelial-mesenchymal transition and GSC maintenance in GBM. Mechanistically, circRYK sponges miR-330-5p and promotes the expression of the oncogene VLDLR. In addition, circRYK could enhance the stability of VLDLR mRNA via the RNA-binding protein HuR. CONCLUSION Our findings show that TGF-β promotes epithelial-mesenchymal transition and GSC maintenance in GBM through the circRYK-VLDLR axis, which may provide a new therapeutic target for the treatment of GBM.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China
| | - Binbin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China
| | - Wenping Cao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China.
| | - Xiupeng Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China.
| |
Collapse
|
39
|
Ji R, Chang L, An C, Zhang J. Proton-sensing ion channels, GPCRs and calcium signaling regulated by them: implications for cancer. Front Cell Dev Biol 2024; 12:1326231. [PMID: 38505262 PMCID: PMC10949864 DOI: 10.3389/fcell.2024.1326231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Extracellular acidification of tumors is common. Through proton-sensing ion channels or proton-sensing G protein-coupled receptors (GPCRs), tumor cells sense extracellular acidification to stimulate a variety of intracellular signaling pathways including the calcium signaling, which consequently exerts global impacts on tumor cells. Proton-sensing ion channels, and proton-sensing GPCRs have natural advantages as drug targets of anticancer therapy. However, they and the calcium signaling regulated by them attracted limited attention as potential targets of anticancer drugs. In the present review, we discuss the progress in studies on proton-sensing ion channels, and proton-sensing GPCRs, especially emphasizing the effects of calcium signaling activated by them on the characteristics of tumors, including proliferation, migration, invasion, metastasis, drug resistance, angiogenesis. In addition, we review the drugs targeting proton-sensing channels or GPCRs that are currently in clinical trials, as well as the relevant potential drugs for cancer treatments, and discuss their future prospects. The present review aims to elucidate the important role of proton-sensing ion channels, GPCRs and calcium signaling regulated by them in cancer initiation and development. This review will promote the development of drugs targeting proton-sensing channels or GPCRs for cancer treatments, effectively taking their unique advantage as anti-cancer drug targets.
Collapse
Affiliation(s)
- Renhui Ji
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Li Chang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Caiyan An
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| | - Junjing Zhang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| |
Collapse
|
40
|
Binobaid L, As Sobeai HM, Alhazzani K, AlAbdi L, Alwazae MM, Alotaibi M, Parrington J, Alhoshani A. Whole-exome sequencing identifies cancer-associated variants of the endo-lysosomal ion transport channels in the Saudi population. Saudi Pharm J 2024; 32:101961. [PMID: 38313820 PMCID: PMC10832475 DOI: 10.1016/j.jsps.2024.101961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2024] Open
Abstract
Background Although national efforts are underway to document the genomic variability of the Saudi population relative to other populations, such variability remains largely unexplored. Genetic variability is known to impact the fate of cells and increase or decrease the risk of a variety of complex diseases including cancer forms. Therefore, the identification of variants associated with cancer susceptibility in Saudi population may protect individuals from cancer or aid in patient-tailored therapies. The endo-lysosomal ion transport genes responsible for cationic ion homeostasis within the cell. We screened 703 single-nucleotide polymorphisms (SNPs) of the endo-lysosomal ion transporter genes in the Saudi population and identified cancer-associated variants that have been reported in other populations. Methods Utilizing previously derived local data of Whole-Exome Sequencing (WES), we examined SNPs of TPCN1, TPCN2, P2RX4, TRPM7, TRPV4, TRPV4, and TRPV6 genes. The SNPs were identified for those genes by our in-house database. We predicted the pathogenicity of these variants using in silico tools CADD, Polyphen-2, SIFT, PrimateAI, and FATHMM-XF. Then, we validated our findings by exploring the genetics database (VarSome, dbSNP NCB, OMIM, ClinVar, Ensembl, and GWAS Catalog) to further link cancer risk. Results The WES database yielded 703 SNPs found in TPCN2, P2RX4, TRPM7, TRPV4, and TRPV6 genes in 1,144 subjects. The number of variants that were found to be common in our population was 150 SNPs. We identified 13 coding-region non-synonymous variants of the endo-lysosomal genes that were most common with a minor allele frequency (MAF) of ≥ 1 %. Twelve of these variants are rs2376558, rs3750965, rs61746574, rs35264875, rs3829241, rs72928978, rs25644, rs8042919, rs17881456, rs4987682, rs4987667, and rs4987657 that were classified as cancer-associated genes. Conclusion Our study highlighted cancer-associated SNPs in the endo-lysosomal genes among Saudi individuals. The allelic frequencies on polymorphic variants confer susceptibility to complex diseases that are comparable to other populations. There is currently insufficient clinical data supporting the link between these SNPs and cancer risk in the Saudi population. Our data argues for initiating future cohort studies in which individuals with the identified SNPs are monitored and assessed for their likelihood of developing malignancies and therapy outcomes.
Collapse
Affiliation(s)
- Lama Binobaid
- Dept. of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11454, Saudi Arabia
| | - Homood M. As Sobeai
- Dept. of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11454, Saudi Arabia
| | - Khalid Alhazzani
- Dept. of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11454, Saudi Arabia
| | - Lama AlAbdi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2457, Riyadh 11454, Saudi Arabia
| | - Meshari M. Alwazae
- Computational Sciences Department, Center of Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Moureq Alotaibi
- Dept. of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11454, Saudi Arabia
| | - John Parrington
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Ali Alhoshani
- Dept. of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11454, Saudi Arabia
| |
Collapse
|
41
|
Jiang Y, Shen L, Wang B. Non-electrophysiological techniques targeting transient receptor potential (TRP) gene of gastrointestinal tract. Int J Biol Macromol 2024; 262:129551. [PMID: 38367416 DOI: 10.1016/j.ijbiomac.2024.129551] [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: 10/25/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 02/19/2024]
Abstract
Transient receptor potential (TRP) channels are cation channels related to a wide range of physical and chemical stimuli, they are expressed all along the gastrointestinal system, and a myriad of diseases are often associated with aberrant expression or mutation of the TRP gene, suggesting that TRPs are promising targets for drug therapy. Therefore, a better understanding of the information of TRPs in health and disease could facilitate the development of effective drugs for the treatment of gastrointestinal diseases like IBD. But there are very few generalizations about the experimental techniques studied in this field. In view of the promise of TRP as a therapeutic target, we discuss experimental methods that can be used for TRPs including their distribution, function and interaction with other proteins, as well as some promising emerging technologies to provide experimental methods for future studies.
Collapse
Affiliation(s)
- Yuting Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Lan Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
42
|
Chen L, Xu YX, Wang YS, Zhou JL. Lipid metabolism, amino acid metabolism, and prostate cancer: a crucial metabolic journey. Asian J Androl 2024; 26:123-134. [PMID: 38157428 PMCID: PMC10919422 DOI: 10.4103/aja202363] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/08/2023] [Indexed: 01/03/2024] Open
Abstract
Prostate cancer (PCa) is one of the most common malignancies in males worldwide, and its development and progression involve the regulation of multiple metabolic pathways. Alterations in lipid metabolism affect the proliferation and metastatic capabilities of PCa cells. Cancer cells increase lipid synthesis and regulate fatty acid oxidation to meet their growth and energy demands. Similarly, changes occur in amino acid metabolism in PCa. Cancer cells exhibit an increased demand for specific amino acids, and they regulate amino acid transport and metabolic pathways to fulfill their proliferation and survival requirements. These changes are closely associated with disease progression and treatment response in PCa cells. Therefore, a comprehensive investigation of the metabolic characteristics of PCa is expected to offer novel insights and approaches for the early diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Yuan-Shuo Wang
- School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Jian-Liang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
43
|
Zong S, Gao J. Identifying the tumor immune microenvironment-associated prognostic genes for prostate cancer. Discov Oncol 2024; 15:42. [PMID: 38376699 PMCID: PMC10879074 DOI: 10.1007/s12672-023-00856-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/29/2023] [Indexed: 02/21/2024] Open
Abstract
PURPOSE This study aimed to explore novel tumor immune microenvironment (TIME)-associated biomarkers in prostate adenocarcinoma (PRAD). METHODS PRAD RNA-sequencing data were obtained from UCSC Xena database as the training dataset. The ESTIMATE package was used to evaluate stromal, immune, and tumor purity scores. Differentially expressed genes (DEGs) related to TIME were screened using the immune and stromal scores. Gene functions were analyzed using DAVID. The LASSO method was performed to screen prognostic TIME-related genes. Kaplan-Meier curves were used to evaluate the prognosis of samples. The correlation between the screened genes and immune cell infiltration was explored using Tumor IMmune Estimation Resource. The GSE70768 dataset from the Gene Expression Omnibus was used to validate the expression of the screened genes. RESULTS The ESTIMATE results revealed that high immune, stromal, and ESTIMATE scores and low tumor purity had better prognoses. Function analysis indicated that DEGs are involved in the cytokine-cytokine receptor interaction signaling pathway. In TIME-related DEGs, METTL7B, HOXB8, and TREM1 were closely related to the prognosis. Samples with low expression levels of METTL7B, HOXB8, and TREM1 had better survival times. Similarly, both the validation dataset and qRT-PCR suggested that METTL7B, HOXB8, and TREM1 were significantly decreased. The three genes showed a positive correlation with immune infiltration. CONCLUSIONS This study identified three TIME-related genes, namely, METTL7B, HOXB8, and TREM1, which correlated with the prognosis of patients with PRAD. Targeting the TIME-related genes might have important clinical implications when making decisions for immunotherapy in PRAD.
Collapse
Affiliation(s)
- Shi Zong
- Department of Urology, Union Hospital of Jilin University, No.126, Xian Tai Road, Chang Chun, 130021, China
| | - Ji Gao
- Department of Urology, Union Hospital of Jilin University, No.126, Xian Tai Road, Chang Chun, 130021, China.
| |
Collapse
|
44
|
Xiong J, Xiao R, Zhao J, Zhao Q, Luo M, Li F, Zhang W, Wu M. Matrix stiffness affects tumor-associated macrophage functional polarization and its potential in tumor therapy. J Transl Med 2024; 22:85. [PMID: 38246995 PMCID: PMC10800063 DOI: 10.1186/s12967-023-04810-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/17/2023] [Indexed: 01/23/2024] Open
Abstract
The extracellular matrix (ECM) plays critical roles in cytoskeletal support, biomechanical transduction and biochemical signal transformation. Tumor-associated macrophage (TAM) function is regulated by matrix stiffness in solid tumors and is often associated with poor prognosis. ECM stiffness-induced mechanical cues can activate cell membrane mechanoreceptors and corresponding mechanotransducers in the cytoplasm, modulating the phenotype of TAMs. Currently, tuning TAM polarization through matrix stiffness-induced mechanical stimulation has received increasing attention, whereas its effect on TAM fate has rarely been summarized. A better understanding of the relationship between matrix stiffness and macrophage function will contribute to the development of new strategies for cancer therapy. In this review, we first introduced the overall relationship between macrophage polarization and matrix stiffness, analyzed the changes in mechanoreceptors and mechanotransducers mediated by matrix stiffness on macrophage function and tumor progression, and finally summarized the effects of targeting ECM stiffness on tumor prognosis to provide insight into this new field.
Collapse
Affiliation(s)
- Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Rourou Xiao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jiahui Zhao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qiuyan Zhao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Manwen Luo
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Feng Li
- Department of Medical Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Allergy and Immunology, Wuhan, 430071, China.
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, China.
| |
Collapse
|
45
|
Köles L, Ribiczey P, Szebeni A, Kádár K, Zelles T, Zsembery Á. The Role of TRPM7 in Oncogenesis. Int J Mol Sci 2024; 25:719. [PMID: 38255793 PMCID: PMC10815510 DOI: 10.3390/ijms25020719] [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: 11/28/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
This review summarizes the current understanding of the role of transient receptor potential melastatin-subfamily member 7 (TRPM7) channels in the pathophysiology of neoplastic diseases. The TRPM family represents the largest and most diverse group in the TRP superfamily. Its subtypes are expressed in virtually all human organs playing a central role in (patho)physiological events. The TRPM7 protein (along with TRPM2 and TRPM6) is unique in that it has kinase activity in addition to the channel function. Numerous studies demonstrate the role of TRPM7 chanzyme in tumorigenesis and in other tumor hallmarks such as proliferation, migration, invasion and metastasis. Here we provide an up-to-date overview about the possible role of TRMP7 in a broad range of malignancies such as tumors of the nervous system, head and neck cancers, malignant neoplasms of the upper gastrointestinal tract, colorectal carcinoma, lung cancer, neoplasms of the urinary system, breast cancer, malignant tumors of the female reproductive organs, prostate cancer and other neoplastic pathologies. Experimental data show that the increased expression and/or function of TRPM7 are observed in most malignant tumor types. Thus, TRPM7 chanzyme may be a promising target in tumor therapy.
Collapse
Affiliation(s)
- László Köles
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Polett Ribiczey
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Andrea Szebeni
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
| | - Kristóf Kádár
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
| | - Tibor Zelles
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, H-1083, Budapest, Hungary
| | - Ákos Zsembery
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary; (L.K.); (A.S.); (K.K.); (T.Z.)
| |
Collapse
|
46
|
Liang H, Yin G, Shi G, Liu Z, Liu X, Li J. Echinacoside regulates PI3K/AKT/HIF-1α/VEGF cross signaling axis in proliferation and apoptosis of breast cancer. Anal Biochem 2024; 684:115360. [PMID: 37865269 DOI: 10.1016/j.ab.2023.115360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
CONTEXT Echinacoside (ECH) is a natural anti-cancer compound and is of great value in cancer treatment. However, the mechanism underlying this effect on breast cancer (BC) was unclear. OBJECTIVE To explore the mechanism of ECH treating BC by network pharmacology and experimental validation. MATERIALS & METHODS Several databases were searched to screen potential targets of ECH and obtain information on targets related to BC. STRING was applied to construct a Protein-protein interaction (PPI) network. DAVID was applied for Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Gene Expression Profiling Interactive Analysis (GEPIA) was searched for the relationship between the expression profile and overall survival of major targets in normal breast and BC tissues. Finally, the results of network pharmacology analysis were validated by experiments. RESULTS Seventeen targets of ECH overlapped with targets in BC. Ten hub targets were determined through PPI. By GO and KEGG analysis 15 entries and 25 pathways were obtained, in which phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT), hypoxia inducible factor-1 (HIF-1) and vascular endothelial growth factor (VEGF) played greater roles. Validation of key targets in the GEPIA database showed that PIK3R1 and PIK3CD remained consistent with the results of the study. Experiments in vitro showed ECH inhibited proliferation, induced apoptosis and reduced mRNA levels and protein expression of PI3K, AKT, hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor A (VEGFA) in MCF-7 cells. Furthermore, experiments in vivo revealed that ECH significantly reduced tumor growth, promoted apoptosis and decreased the related mRNA levels and protein expression, suggesting ECH works on BC by regulating PI3K/AKT/HIF-1α/VEGF signaling pathway. DISCUSSION & CONCLUSION In summary, ECH played an important role in anti-BC by regulating PI3K/AKT/HIF-1α/VEGF signaling pathway. Furthermore, ECH had multi-target and multi-pathway effects, which may be a promising natural compound for treating BC.
Collapse
Affiliation(s)
- Hongyi Liang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Guangxi Shi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Zhiyong Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Xiaofei Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Jingwei Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
47
|
Zou W, Zhang L, Hu Y, Gao Y, Zhang J, Zheng J. The role of TRPV ion channels in adipocyte differentiation: What is the evidence? Cell Biochem Funct 2024; 42:e3933. [PMID: 38269518 DOI: 10.1002/cbf.3933] [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: 07/25/2023] [Revised: 12/27/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024]
Abstract
Obesity is a complex disorder, and the incidence of obesity continues to rise at an alarming rate worldwide. In particular, the growing incidence of overweight and obesity in children is a major health concern. However, the underlying mechanisms of obesity remain unclear and the efficacy of several approaches for weight loss is limited. As an important calcium-permeable temperature-sensitive cation channel, transient receptor potential vanilloid (TRPV) ion channels directly participate in thermo-, mechano-, and chemosensory responses. Modulation of TRPV ion channel activity can alter the physiological function of the ion channel, leading to neurodegenerative diseases, chronic pain, cancer, and skin disorders. In recent years, increasing studies have demonstrated that TRPV ion channels are abundantly expressed in metabolic organs, including the liver, adipose tissue, skeletal muscle, pancreas, and central nervous system, which has been implicated in various metabolic diseases, including obesity and diabetes mellitus. In addition, as an important process for the pathophysiology of adipocyte metabolism, adipocyte differentiation plays a critical role in obesity. In this review, we focus on the role of TRPV ion channels in adipocyte differentiation to broaden the ideas for prevention and control strategies for obesity.
Collapse
Affiliation(s)
- Wenyu Zou
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ling Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Yongyan Hu
- Laboratory Animal Facility, Peking University First Hospital, Beijing, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| |
Collapse
|
48
|
Mei XC, Chen Q, Zuo S. Transient receptor potential-related risk model predicts prognosis of hepatocellular carcinoma patients. World J Gastrointest Oncol 2023; 15:2064-2076. [PMID: 38173438 PMCID: PMC10758653 DOI: 10.4251/wjgo.v15.i12.2064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/17/2023] [Accepted: 11/10/2023] [Indexed: 12/14/2023] Open
Abstract
BACKGROUND Members of the transient receptor potential (TRP) protein family shape oncogenic development, but the specific relevance of TRP-related genes in hepatocellular carcinoma (HCC) has yet to be defined. AIM To investigate the role of TRP genes in HCC, their association with HCC development and treatment was examined. METHODS HCC patient gene expression and clinical data were downloaded from The Cancer Genome Atlas database, and univariate and least absolute shrinkage and selection operator Cox regression models were employed to explore the TRP-related risk spectrum. Based on these analyses, clinically relevant TRP family genes were selected, and the association between the key TRP canonical type 1 (TRPC1) gene and HCC patient prognosis was evaluated. RESULTS In total, 28 TRP family genes were screened for clinical relevance, with multivariate analyses ultimately revealing three of these genes (TRPC1, TRP cation channel subfamily M member 2, and TRP cation channel subfamily M member 6) to be significantly associated with HCC patient prognosis (P < 0.05). These genes were utilized to establish a TRP-related risk model. Patients were separated into low- and high-risk groups based on the expression of these genes, and high-risk patients exhibited a significantly poorer prognosis (P = 0.001). Functional analyses highlighted pronounced differences in the immune status of patients in these two groups and associated enriched immune pathways. TRPC1 was identified as a candidate gene in this family worthy of further study, with HCC patients expressing higher TRPC1 levels exhibiting poorer survival outcomes. Consistently, quantitative, immunohistochemistry, and western blot analyses revealed increased TRPC1 expression in HCC. CONCLUSION These three TRP genes help determine HCC patient prognosis, providing insight into tumor immune status and immunological composition. These findings will help design combination therapies including immunotherapeutic and anti-TRP agents.
Collapse
Affiliation(s)
- Xiao-Cai Mei
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, Guizhou Province, China
| | - Qian Chen
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, Guizhou Province, China
| | - Shi Zuo
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, Guizhou Province, China
| |
Collapse
|
49
|
Chen W, Xu Z, Jiang J, Chen L, Chen Y, Yu T, Chen H, Shi R. CXCL8 as a Potential Biomarker for Crohn's Disease is Associated with Immune Infiltration and RNA Methylation. Biochem Genet 2023; 61:2599-2617. [PMID: 37202601 DOI: 10.1007/s10528-023-10397-7] [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: 03/02/2023] [Accepted: 05/02/2023] [Indexed: 05/20/2023]
Abstract
TRP channels have an important role in regulating the function of gastrointestinal epithelial cells. The aim of this study was to investigate the molecular mechanisms of genes associated with TRP channels in Crohn's disease (CD) by bioinformatics approach and to identify potential key biomarkers. In our study, we identified TRP channel-related differentially expressed genes (DEGs) based on the GSE95095 dataset and the TRP channel-related gene set from the GeneCards database. Hub genes (CXCL8, HIF1A, NGF, JUN, IL1A) were identified by the PPI network and validated by the external GSE52746 dataset. Immune infiltration analysis revealed that CXCL8 was significantly correlated with B cells memory, NK cells activated, Mast cells resting, Mast cells activated, and Neutrophils. GSEA of CXCL8 results showed inositol phosphate metabolism, RNA polymerase, propanoate metabolism, MAPK signaling pathway, base excision repair, and Calcium signaling pathway. In addition, we constructed a lncRNA-miRNA-mRNA ceRNA network and a drug-gene interaction network. Finally, we performed in vitro experiments to verify that LPS induced CXCL8 expression in HT-29 cells and that knockdown of CXCL8 inhibited the inflammatory stimulatory effects of LPS. This study reveals that CXCL8 plays an important role in the pathogenesis of Crohn's disease and is expected to be a novel biomarker.
Collapse
Affiliation(s)
- Wei Chen
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Zeyan Xu
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Jingjing Jiang
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Lu Chen
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Yanfang Chen
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Ting Yu
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Hong Chen
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| | - Ruihua Shi
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
50
|
Ghafouri E, Bigdeli M, Khalafiyan A, Amirkhani Z, Ghanbari R, Hasan A, Khanahmad H, Boshtam M, Makvandi P. Unmasking the complex roles of hypocalcemia in cancer, COVID-19, and sepsis: Engineered nanodelivery and diagnosis. ENVIRONMENTAL RESEARCH 2023; 238:116979. [PMID: 37660871 DOI: 10.1016/j.envres.2023.116979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/05/2023]
Abstract
Calcium (Ca2+) homeostasis is essential for maintaining physiological processes in the body. Disruptions in Ca2+ signaling can lead to various pathological conditions including inflammation, fibrosis, impaired immune function, and accelerated senescence. Hypocalcemia, a common symptom in diseases such as acute respiratory distress syndrome (ARDS), cancer, septic shock, and COVID-19, can have both potential protective and detrimental effects. This article explores the multifaceted role of Ca2+ dysregulation in inflammation, fibrosis, impaired immune function, and accelerated senescence, contributing to disease severity. Targeting Ca2+ signaling pathways may provide opportunities to develop novel therapeutics for age-related diseases and combat viral infections. However, the role of Ca2+ in viral infections is complex, and evidence suggests that hypocalcemia may have a protective effect against certain viruses, while changes in Ca2+ homeostasis can influence susceptibility to viral infections. The effectiveness and safety of Ca2+ supplements in COVID-19 patients remain a subject of ongoing research and debate. Further investigations are needed to understand the intricate interplay between Ca2+ signaling and disease pathogenesis.
Collapse
Affiliation(s)
- Elham Ghafouri
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Anis Khalafiyan
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zohre Amirkhani
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roham Ghanbari
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| |
Collapse
|