1
|
Merz LM, Winter K, Richter S, Kallendrusch S, Horn A, Grunewald S, Klöting N, Krause K, Kiess W, Le Duc D, Garten A. Effects of alpelisib treatment on murine Pten-deficient lipomas. Adipocyte 2025; 14:2468275. [PMID: 39962643 PMCID: PMC11844927 DOI: 10.1080/21623945.2025.2468275] [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: 09/25/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/23/2025] Open
Abstract
Phosphatase and tensin homolog (PTEN) hamartoma tumour syndrome (PHTS) is a rare disorder caused by germline mutations in the tumour suppressor gene PTEN, a key negative regulator of phosphatidylinositol 3-kinase (PI3K)/AKT signalling. Children with PHTS often develop lipomas, for which only surgical resection is available as treatment. We investigated the effects of the selective PI3K-inhibitor alpelisib on Pten-deficient lipomas. After incubation with alpelisib or the non-selective PI3K inhibitor wortmannin, we analysed histology, gene expression, and Pi3k pathway in lipoma and control epididymal adipose tissue (epiWAT). Alpelisib increased adipocyte area in lipomas compared to epiWAT. Baseline gene expression showed higher levels of markers for proliferation (Pcna), fibrosis (Tgfb1), and adipogenesis (Pparg) in lipomas, while hormone-sensitive lipase expression was lower than in epiWAT. Following alpelisib incubation, target genes of Pi3k signalling and extracellular matrix factors were reduced. We confirmed Pi3k inhibition through detecting decreased Akt levels compared to control treatment. Human lipoma samples treated with alpelisib showed variable lipolysis responses, suggesting variability in therapeutic outcomes. We established an ex vivo model to study alpelisib effects on Pten-deficient lipomas. These results underscore the therapeutic potential of targeted PI3K inhibition in the treatment of PHTS-associated lipomas, particularly in cases that are inoperable.
Collapse
Affiliation(s)
- Lea M. Merz
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| | - Karsten Winter
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Sandy Richter
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| | - Sonja Kallendrusch
- Institute of Anatomy, Leipzig University, Leipzig, Germany
- Institute of Clinical Research and Systems Medicine, Health and Medical University Potsdam, Potsdam, Germany
| | - Andreas Horn
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Sonja Grunewald
- Department for Dermatology, Venereology and Allergology, University Hospital Leipzig, Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Center Munich at the University and University Hospital Leipzig, Leipzig, Germany
| | - Kerstin Krause
- Department of Endocrinology, Nephrology and Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Wieland Kiess
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| | - Diana Le Duc
- Institute of Human Genetics, University Hospital Leipzig, Leipzig, Germany
| | - Antje Garten
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| |
Collapse
|
2
|
Han Y, Sun Y, Peng S, Tang T, Zhang B, Yu R, Sun X, Guo S, Ma L, Li P, Yang P. PI3K/AKT pathway: A potential therapeutic target in cerebral ischemia-reperfusion injury. Eur J Pharmacol 2025; 998:177505. [PMID: 40118329 DOI: 10.1016/j.ejphar.2025.177505] [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: 10/02/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
Cerebral ischemia is a prevalent cerebrovascular disorder, with the restoration of blocked blood vessels serving as the current standard clinical treatment. However, reperfusion can exacerbate neuronal damage and neurological dysfunction, resulting in cerebral ischemia-reperfusion (I/R) injury. Presently, clinical treatment strategies for cerebral I/R injury are limited, creating an urgent need to identify new effective therapeutic targets. The PI3K/AKT signaling pathway, a pro-survival pathway associated with cerebral I/R injury, has garnered significant attention. We conducted a comprehensive review of the literature on the PI3K/AKT pathway in the context of cerebral I/R. Our findings indicate that activation of the PI3K/AKT signaling pathway following cerebral I/R can alleviate oxidative stress, reduce endoplasmic reticulum stress (ERS), inhibit inflammatory responses, decrease neuronal apoptosis, autophagy, and pyroptosis, mitigate blood-brain barrier (BBB) damage, and promote neurological function recovery. Consequently, this pathway ultimately reduces neuronal death, alleviates brain tissue damage, decreases the volume of cerebral infarction, and improves behavioral impairments. These results suggest that the PI3K/AKT signaling pathway is a promising therapeutic target for further research and drug development, holding significant potential for the treatment of cerebral I/R injury.
Collapse
Affiliation(s)
- Yiming Han
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yu Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shiyu Peng
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Tingting Tang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Beibei Zhang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Ruonan Yu
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Xiaoyan Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shanshan Guo
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China; Staff Hospital of Henan Fifth Construction Group Co., Ltd, Zhengzhou, Henan, China
| | - Lijuan Ma
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Pengfei Yang
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| |
Collapse
|
3
|
Peng L, Rahman Z, Tian Y, Yin T, Xiong S, You J, Liu R, Wang L, Huang Q, Ma H. Comprehensive molecular biology and metabolomics analysis reveal the changes on muscle quality of Megalobrama amblycephala exposure to ammonia nitrogen during transportation. Food Res Int 2025; 212:116372. [PMID: 40382029 DOI: 10.1016/j.foodres.2025.116372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/23/2025] [Accepted: 04/15/2025] [Indexed: 05/20/2025]
Abstract
This study comprehensively investigated the effects of different ammonia nitrogen models during transportation on the energy metabolism, redox system, apoptosis, and changes in muscle quality of fish using molecular biology and metabolomics. Exposure to ammonia nitrogen caused intensive stress response as evidenced by alteration on the levels of biochemical indicators (cortisol, glucose, urea nitrogen, alanine transaminase, lactic dehydrogenase, superoxide dismutase, and glutathione peroxidase) and structural disruption of organs (including gill, cephalic kidney, kidney, and liver). As a result of the ammonia nitrogen stress, the redox system became imbalance, leading to disturbance in energy metabolism primarily through the pathways of D-amino acid metabolism, alanine/aspartate/glutamate metabolism, and purine metabolism. Additionally, apoptosis occurred following stress, regulated by FoxO, mTOR, NF-κB, and PI3K/AKT signaling pathways. Besides redox system, energy metabolism, and apoptosis, the change of muscle quality were also influenced by ammonia nitrogen concentration and exposure duration. Drip loss increased with higher ammonia nitrogen concentrations and longer exposure time, while shear force value showed an inverse trend. Although no significant changes were observed in a* and b* values following ammonia nitrogen exposure, the highest W and L* values were found in the low-concentration groups. The correlation of spearman indicates the changes in muscle quality, including drip loss, shear force, and color, induced by ammonia nitrogen during transportation was attributed to the interplay of redox system, energy metabolism, and apoptosis.
Collapse
Affiliation(s)
- Ling Peng
- College of Food Science and Technology/National R&D Branch Center for Conventional Freshwater Fish Processing (Wuhan), Huazhong Agricultural University, Wuhan 430070, PR China
| | - Ziaur Rahman
- College of Food Science and Technology/National R&D Branch Center for Conventional Freshwater Fish Processing (Wuhan), Huazhong Agricultural University, Wuhan 430070, PR China
| | - Yuanyong Tian
- College of Food Science and Engineering, Dalian Ocean University 116023, PR China
| | - Tao Yin
- College of Food Science and Technology/National R&D Branch Center for Conventional Freshwater Fish Processing (Wuhan), Huazhong Agricultural University, Wuhan 430070, PR China.
| | - Shanbai Xiong
- College of Food Science and Technology/National R&D Branch Center for Conventional Freshwater Fish Processing (Wuhan), Huazhong Agricultural University, Wuhan 430070, PR China
| | - Juan You
- College of Food Science and Technology/National R&D Branch Center for Conventional Freshwater Fish Processing (Wuhan), Huazhong Agricultural University, Wuhan 430070, PR China
| | - Ru Liu
- College of Food Science and Technology/National R&D Branch Center for Conventional Freshwater Fish Processing (Wuhan), Huazhong Agricultural University, Wuhan 430070, PR China
| | - Lan Wang
- Institute of Agricultural Products Processing and Nuclear Agricultural Technology, Hubei Academy of Agricultural Sciences, Wuhan 430064, PR China
| | - Qilin Huang
- College of Food Science and Technology/National R&D Branch Center for Conventional Freshwater Fish Processing (Wuhan), Huazhong Agricultural University, Wuhan 430070, PR China
| | - Huawei Ma
- Engineering Research Center of Processing & Storage of Characteristic and Advantage Aquatic Products from Guangxi, Guangxi Academy of Fishery Science, Nanning 530021, Guangxi, China
| |
Collapse
|
4
|
Cai J, Zhou R, Ye Y, Hao J, Luo Q, Mei H, He Z, Wang F, Yalikun A, Yu Y, Wen Y. UGDH promotes 5-fluorouracil resistance in colorectal cancer via the ROS-activated PI3K/AKT-EEF1A2-PRDX1 pathway. Arch Biochem Biophys 2025; 769:110445. [PMID: 40311992 DOI: 10.1016/j.abb.2025.110445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 04/01/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
5-Fluorouracil (5-FU) chemotherapy resistance is a critical determinant of poor prognosis in patients with colorectal cancer (CRC). One critical mechanism underlying this resistance is the clearance of reactive oxygen species (ROS) generated by 5-FU, which diminishes its cytotoxic efficacy. Here, we identified the differential expression of UDP-glucose dehydrogenase (UGDH) in resistant cells through sequencing, and downstream targets EEF1A2 and PRDX1 were identified via immunoprecipitation-mass spectrometry (IP-MS). Stable knockdown and overexpression cell models were generated using a lentiviral system. The effects of gene manipulation on 5-FU resistance in CRC were evaluated both in vitro and in vivo through flow cytometry for reactive oxygen species (ROS) and apoptosis, as well as TUNEL immunofluorescence assays. Sequencing was utilized to enrich the relevant pathways. Our study firstly demonstrates that ROS-induced activation of the PI3K/AKT signaling pathway upregulates UGDH expression. UGDH promotes 5-FU resistance by collaborating with downstream effectors EEF1A2 and PRDX1 to clear ROS and inhibit tumor cell apoptosis. UGDH serves as a potential biomarker for 5-FU resistance in CRC, with its expression levels providing a crucial basis for therapeutic decision-making.
Collapse
Affiliation(s)
- Jinfeng Cai
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runkai Zhou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingzi Ye
- Department of Infectious Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Jialing Hao
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - Qinshan Luo
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haitao Mei
- Department of Gastrointestinal Surgery, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Middle Rd., Shanghai, 200071, China
| | - Zeping He
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fazhi Wang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Abudushalamu Yalikun
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Yu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yugang Wen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Arabi K, Nazemi Salman B, Rahimzadeh-Bajgiran F, Moghbeli M, Moghadas S, Saburi E. miRNAs in oral cancer; diagnostic and prognostic roles. Gene 2025; 951:149382. [PMID: 40049425 DOI: 10.1016/j.gene.2025.149382] [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: 11/23/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
Oral cancer (OC) has become increasingly prevalent in recent years, making it one of the most often occurring types of cancer in patients. The clinical identification of OC is usually a time-consuming procedure, and the outlook for individuals with OC is generally unfavorable, as no particular biomarkers have been established to far. The main risk factors linked to OC are high levels of tobacco and alcohol intake, together with a reduced occurrence of viral infections, such as human papillomavirus. Furthermore, there is evidence suggesting that genetic characteristics that can be passed down from parents to offspring play a role in increasing the likelihood of getting ovarian cancer. MicroRNAs (miRNAs) are brief RNA molecules that do not code for proteins and have the ability to either repress or promote the growth of tumors during cancer development. They have been discovered to control multiple signaling pathways within cells, and their abnormal regulation has been demonstrated to be crucial in initiating and furthering the development of cancer. Additionally, they have the ability to either facilitate or impede the entire multi-stage process of cancer metastasis, including epithelial-mesenchymal transition (EMT), migration, and invasion, by selectively targeting essential genes involved in these pathways. Several microRNAs have the ability to regulate gene expression through various ways. In addition, like other types of cancer, OC has shown alterations in the expression of miRNAs, and certain miRNAs may have the ability to be used for diagnosis and treatment. The investigation of these miRNA could perhaps result in advancements in the specified instances of OC.
Collapse
Affiliation(s)
- Kimia Arabi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Bahareh Nazemi Salman
- Department of Pediatric Dentistry, School of Dentistry, Zanjan University of Medical Sciences, Zanjan 4513956184, Iran.
| | | | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sepehr Moghadas
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Ehsan Saburi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Wan G, Li Z, Gu L, Sun Y, Wang Y, Wang Y, Geng R, Chen Y, Ma W, Bao X, Wang R. Endoscopic nasal delivery of engineered endothelial progenitor cell-derived exosomes improves angiogenesis and neurological deficits in rats with intracerebral hemorrhage. Mater Today Bio 2025; 32:101652. [PMID: 40160244 PMCID: PMC11953990 DOI: 10.1016/j.mtbio.2025.101652] [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: 11/23/2024] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025] Open
Abstract
Intracerebral hemorrhage (ICH) remains a life-threatening condition due to its high mortality and limited treatment options. This study explores a novel therapeutic strategy using engineered exosomes derived from endothelial progenitor cells (EPC-EXOs) to improve ICH outcomes. EPC-EXOs were modified with a CD47-enriched red blood cell membrane via co-extrusion to enhance their anti-phagocytic properties, thereby reducing degradation by activated microglia after ICH. A minimally invasive endoscopic-guided delivery system was developed to facilitate the targeted intranasal administration of these engineered EPC-EXOs (m-Oe-EXOs), allowing direct entry into brain tissue. We confirmed m-Oe-EXOs' high retention and effective distribution in the brain. Functional analysis demonstrated that EPC-EXOs significantly promoted the proliferation, migration, and angiogenesis of brain microvascular endothelial cells (BMECs), with proteomic analysis identifying HSP90 as a key protein activating the Akt pathway in BMECs. In vivo, m-Oe-EXOs demonstrated therapeutic efficacy by improving blood-brain barrier integrity, reducing hematoma volume, and enhancing neurological recovery in ICH rats. Collectively, our findings highlight the potential of minimally invasive, endoscopic-guided delivery of m-Oe-EXOs as an innovative approach for ICH treatment, providing new insights into targeted, exosomes-based regenerative therapies.
Collapse
Affiliation(s)
- Gui Wan
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhenwei Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lingui Gu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ye Sun
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yuhe Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yiqing Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruxu Geng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yangyang Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical College, Hefei, 230031, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| |
Collapse
|
7
|
Liu Y, Wang X, Li Z, Gao X, Wu X, Pi J, Wang X, Wang Q, Zhou F, Wang X. Melatonin attenuates brain edema via the PI3K/Akt/Nrf2 pathway in rats with cerebral ischemia-reperfusion injury. J Stroke Cerebrovasc Dis 2025; 34:108299. [PMID: 40158783 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 03/09/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
OBJECTIVE This study aimed to explore the neuroprotective effects of Melatonin (Mel) administration on cerebral ischemia-reperfusion injury (CIRI) and elucidate its underlying mechanism in vivo to provide a theoretical foundation for the clinical application of Mel. MATERIALS AND METHODS CIRI models were established in male adult Sprague Dawley rats by middle cerebral artery occlusion (MCAO) for 2 h. Water content of brain tissue was assessed using both dry/wet weight method and T2-weighted Imaging (T2WI). The infarct volume of the brain was measured by 2,3,5-triphenyltetrazolium chloride (TTC) staining. Cell morphology changes and brain damage were detected through hematoxylin & eosin (H&E) staining and NeuN immunofluorescence staining. The integrity of blood-brain barrier (BBB) was examined using transmission electron microscopy (TEM). The expression of aquaporin 4 (AQP4) protein was quantified through western blots analysis and immunofluorescence staining. The expression of p-PI3K, p-AKT and Nrf2 proteins were detected by immunohistochemistry staining and western blots analysis. RESULTS Compared with the CIRI group, Mel administration significantly reduced the infarct volume and ameliorated the morphology alterations, accompanied by an increase in the number of neurons. The water content of brain tissue decreased significantly, and the value of relative average diffusion coefficient (rADC) of injured brain increased in the CIRI + Mel group as compared with the CIRI group. Compared with the CIRI group, Mel administration improved the damage to the tight junctions of endothelial cells in the cerebral cortex. The expression of AQP4 protein decreased, and that of p-PI3K, p-AKT and Nrf2 proteins increased in the CIRI + Mel group compared with the CIRI group. After administration of p-PI3K inhibitor LY294002, the expression of AQP4 was upregulated, and that of the p-PI3K, p-AKT and Nrf2 proteins decreased compared with the CIRI + Mel group. CONCLUSIONS Mel administration exerts neuroprotective effects against CIRI by mitigating brain edema through upregulating the PI3K/AKT/Nrf2 signaling pathway, and then attenuating brain damage in CIRI rats.
Collapse
Affiliation(s)
- Yang Liu
- School of Medical Imaging, Shandong Second Medical University, Weifang, 261053, China; Medical Imaging Center, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Xin Wang
- School of Medical Imaging, Shandong Second Medical University, Weifang, 261053, China; Medical Imaging Center, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Zhen Li
- School of Medical Imaging, Shandong Second Medical University, Weifang, 261053, China; Medical Imaging Center, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Xiaotian Gao
- School of Medical Imaging, Shandong Second Medical University, Weifang, 261053, China
| | - Xiaoli Wu
- Department of Radiology, Weifang People's Hospital, Weifang 261041, China
| | - Jiayang Pi
- School of Medical Imaging, Shandong Second Medical University, Weifang, 261053, China; Medical Imaging Center, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Xizhen Wang
- Medical Imaging Center, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China
| | - Qi Wang
- Department of Radiology, Weifang People's Hospital, Weifang 261041, China
| | - Fenghua Zhou
- Department of Pathology, School of Basic Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Xiaoli Wang
- School of Medical Imaging, Shandong Second Medical University, Weifang, 261053, China; Medical Imaging Center, Affiliated Hospital of Shandong Second Medical University, Weifang, 261031, China.
| |
Collapse
|
8
|
Wang P, Ma Y, Rao X, Luo Q, Xiao X, Wang T, Long F. Kaempferol targets Src to exert its chemopreventive effects on mammary tumorigenesis via regulation of the PI3K/AKT pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156701. [PMID: 40220416 DOI: 10.1016/j.phymed.2025.156701] [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: 09/20/2024] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Breast cancer (BC) is a prevalent malignancy that poses significant risks to the health of women worldwide. The incidence and mortality rates of BC continue to be high, despite improvements in diagnosis and treatment, indicating a need for novel prevention strategies. Kaempferol (KAM) is a common dietary flavonoid with known antitumour properties, but its role in the chemoprevention of BC and the underlying mechanisms largely unexplored. PURPOSE This study aimed to evaluate the chemopreventive effects of KAM on carcinogen-induced BC in vivo and in vitro and to elucidate the underlying molecular mechanisms. METHODS In this study, we used an N-methyl-N-nitrosourea (NMU)-induced rat model of BC and 17β-oestradiol (E2)-treated MCF-10A cells to evaluate the chemopreventive effects of KAM on mammary tumorigenesis. The antioxidant capacity of KAM was assessed by measuring oxidative damage marker levels and antioxidant enzyme expression. Flow cytometry and Hoechst 33258 staining were utilized to analyse cell cycle distribution and apoptosis. The core target of KAM was identified by network pharmacology and validated by molecular docking, MD simulation, CESTA, and BLI. KEGG enrichment analysis, molecular biology tests and the application of specific protein inhibitors were conducted to elucidate the molecular mechanisms modulated by KAM. RESULTS In vivo, KAM inhibited the progression of mammary tumours and delayed pathological changes in the morphological structure of mammary gland cells to varying degrees. In vitro, KAM reduced cell viability, migration, and anchorage-independent growth while triggering cell cycle arrest and apoptosis in E2-treated MCF-10A cells. Furthermore, KAM increased cellular antioxidant capacity and attenuated E2-induced oxidative stress. Mechanistically, KAM directly interacted with Src and inhibited its phosphorylation, thus leading to PI3K/AKT pathway inhibition. Notably, the inhibition of E2-induced cell migration and anchorage-independent growth in vitro by Src- or PI3K/AKT pathway-specific inhibitors was not further enhanced when the cells were cultured with KAM. CONCLUSION In summary, KAM targets the Src-mediated PI3K/AKT pathway to reduce oxidative stress and facilitate apoptosis and cell cycle arrest, thereby inhibiting mammary tumorigenesis. Our study is the first to identify Src kinase as a direct target of KAM in mammary tumorigenesis. These findings give significant perspectives on the potential application of KAM in BC chemoprevention.
Collapse
Affiliation(s)
- Pinghan Wang
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China
| | - Yu Ma
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Xiaohui Rao
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Qianwen Luo
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China
| | - Xiao Xiao
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China
| | - Ting Wang
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China.
| | - Fangyi Long
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610032, China.
| |
Collapse
|
9
|
Arghidash F, Gheybi F, Gholamhosseinian H, Kesharwani P, Sahebkar A. Radioprotective and radiosensitizing properties of silymarin/silibinin in response to ionizing radiation. Pathol Res Pract 2025; 270:156002. [PMID: 40347920 DOI: 10.1016/j.prp.2025.156002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 05/06/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Cancer is a health and treatment challenge that the world is facing, and many efforts are being made to develop treatment solutions for all forms of cancer. Radiotherapy (RT), one of the cancer treatment methods, can cause toxicity in healthy cells, even though it has positive effects on killing cancer cells. It is possible for cancer cells to develop resistance to radiotherapy. To address these issues, it can be beneficial to combine treatments. Combining plants with conventional cancer treatment is a viable option, and their potential can be utilized in this area. The therapeutic properties of silymarin and its active ingredient silibinin have been used in traditional medicine for a long time. The purpose of this review is to investigate the radioprotective and radio-sensitizing properties of silymarin/silibinin in cancer treatment.
Collapse
Affiliation(s)
- Faezeh Arghidash
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Gheybi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India; University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Teng Y, Xue H, Deng X, Luo Y, Wu T. The role of phosphatidylethanolamine-binding protein (PEBP) family in various diseases: Mechanisms and therapeutic potential. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:102-113. [PMID: 40220872 DOI: 10.1016/j.pbiomolbio.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
This article focuses on the phosphatidylethanolamine-binding protein (PEBP) family proteins, detailing PEBP1 and PEBP4 due to limited information on PEBP2 and PEBP3, in cellular signaling pathways and research in a spectrum of pathologies, including diverse cancers, metabolic disorders, immunological diseases and a subset of organ-specific diseases. It outlines the mechanisms through which PEBP1 and PEBP4 regulate essential signaling pathways that are critical for cellular processes such as proliferation, apoptosis, and metastasis. Recent advancements have shown further understanding of these proteins' roles in pathophysiology and their potential as future therapeutic targets. The findings suggest that the impact of PEBP1 and PEBP4 on the course of different diseases has underscored their potential for more in-depth medical research and novel clinically targeted therapies.
Collapse
Affiliation(s)
- Yeying Teng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haiping Xue
- Industrial Development Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoliang Deng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yanqun Luo
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
11
|
Shi W, Dong J, Zhong B, Hu X, Zhao C. Predicting the Prognosis of Bladder Cancer Patients Through Integrated Multi-omics Exploration of Chemotherapy-Related Hypoxia Genes. Mol Biotechnol 2025; 67:2367-2381. [PMID: 38806990 PMCID: PMC12055635 DOI: 10.1007/s12033-024-01203-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Bladder cancer is a prevalent malignancy with high mortality rates worldwide. Hypoxia is a critical factor in the development and progression of cancers. However, whether and how hypoxia-related genes (HRGs) could affect the development and the chemotherapy response of bladder cancer is still largely unexplored. This study comprehensively explored the complex molecular landscape associated with hypoxia in bladder cancer by analyzing 260 hypoxia genes based on transcriptomic and genomic data in 411 samples. Employing the 109 dysregulated hypoxia genes for consensus clustering, we delineated two distinct bladder cancer clusters characterized by disparate survival outcomes and distinct oncogenic roles. We defined a HPscore that was correlated with a variety of clinical features, including TNM stages and pathologic grades. Tumor immune landscape analysis identified three immune clusters and close interactions between hypoxia genes and the various immune cells. Utilizing a network-based method, we defined 129 HRGs exerting influence on apoptotic processes and critical signaling pathways in cancer. Further analysis of chemotherapy drug sensitivity identified potential drug-target HRGs. We developed a Risk Score model that was related to the overall survival of bladder cancer patients based on doxorubicin-target HRGs: ACTG2, MYC, PDGFRB, DHRS2, and KLRD1. This study not only enhanced our understanding of bladder cancer at the molecular level but also provided promising avenues for the development of targeted therapies, representing a significant step toward the identification of effective treatments and addressing the urgent need for advancements in bladder cancer management.
Collapse
Affiliation(s)
- Wensheng Shi
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, Hunan, China
- Furong Laboratory, Changsha, 410008, Hunan, China
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jiaming Dong
- Department of Radiation, Cangzhou Central Hospital, Hebei, 061000, China
| | - Bowen Zhong
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, Hunan, China
- Furong Laboratory, Changsha, 410008, Hunan, China
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiheng Hu
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410008, Hunan, China
- Furong Laboratory, Changsha, 410008, Hunan, China
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Chunguang Zhao
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
12
|
Han Z, Wen L. G-quadruplex in cancer energy metabolism: A potential therapeutic target. Biochim Biophys Acta Gen Subj 2025; 1869:130810. [PMID: 40254103 DOI: 10.1016/j.bbagen.2025.130810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
In recent years, energy metabolism in cancer has received increasing attention as an important component of tumor biology, and the functions of transcription factors, mitochondria, reactive oxygen species (ROS) and the autophagy-lysosome system in which have been elucidated. G-quadruplex (G4) is a molecular switch that regulates gene transcription or translation. As an anticancer target, the effect of G4 on cancer cell proliferation, apoptosis, cycle and autophagy has been recognized. The energy metabolism system is a unified whole composed of transcription factors, metabolic regulators, metabolites and signaling pathways that run through the entire cancer process. However, the role of G4 in this complex metabolic network has not been systematically elucidated. In this review, we analyze the close correlation between G4 and transcription factors, mitochondria, ROS and the autophagy-lysosome system and suggest that G4 can exert a marked effect on cancer energy metabolism by regulating the above mentioned key regulatory elements. The anticancer effects of some G4 ligands through regulation of energy metabolism have also been summarized, confirming the clear involvement of G4 in energy metabolism. Although much more research is needed, we propose that G4 may play a critical role in the complex energy metabolism system of cancer, which is a promising target for anticancer strategies focusing on energy metabolism.
Collapse
Affiliation(s)
- Zongqiang Han
- Department of Laboratory Medicine, Beijing Xiaotangshan Hospital, Beijing 102211, China
| | - Lina Wen
- Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.
| |
Collapse
|
13
|
Nayak SPRR, Das A, Ramamurthy K, Pasupuleti M, Rajagopal R, Arockiaraj J. Exposure to bisphenol A and sodium nitrate found in processed meat induces endocrine disruption and dyslipidemia through PI3K/AKT/SREBP pathway in zebrafish larvae. J Nutr Biochem 2025; 140:109887. [PMID: 40023200 DOI: 10.1016/j.jnutbio.2025.109887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Meat is a staple in many cultural diets, and the consumption of processed meats has increased significantly worldwide. The widespread use of sodium nitrate (NaNO3) as a preservative and the unintentional leaching of bisphenol A (BPA) from packaging into meats have raised health concerns. This study evaluates the combined toxicity of BPA and NaNO3 despite their individual safety assessments. Our findings reveal that coexposure to BPA and NaNO3 at levels found in processed meats induces mortality and malformations in zebrafish larvae. The combined exposure triggers oxidative stress, lipid peroxidation, dyslipidemia, inflammation, and apoptosis. Network toxicology analysis elucidates the molecular mechanisms underlying metabolic dysfunction caused by these substances. Dysregulation of genes related to thyroid function (tsh-β, dio-1, thr-b) and inflammation (tnf-α, il-1β, il-6, nfκb) was observed in the co-exposure group. Additionally, this group exhibited increased lipid accumulation, elevated cholesterol and triglyceride levels, and dysregulation of essential lipid metabolism genes (srebp2, pcsk9). Co-exposure also impaired larval motility and behavior, evidenced by hypolocomotion and reduced acetylcholinesterase levels. Further gene expression analysis showed increased levels of pi3k and akt, two major signaling molecules. Ultimately, the simultaneous exposure to BPA and NaNO3 leads to disruptions in the endocrine system and abnormal lipid levels via activating the PI3K/AKT/SREBP pathway.
Collapse
Affiliation(s)
- Santosh Pushpa Ramya Ranjan Nayak
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Anamika Das
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Karthikeyan Ramamurthy
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Mukesh Pasupuleti
- Division of Molecular Microbiology & Immunology, CSIR-Central Drug Research Institute (CDRI), Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajakrishnan Rajagopal
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
14
|
Chakravarti B, Tomar MS, Qais FA, Raza S, Abdullah KM, Sharma G, Tewari A, Yadav A, Gupta P, Chattopadhyay N, Shrivastava A, Sinha RA, Siddiqui JA. Alpha lipoic acid modulates metabolic reprogramming in breast cancer stem cells enriched 3D spheroids by targeting phosphoinositide 3-kinase: In silico and in vitro insights. Biomed Pharmacother 2025; 187:118121. [PMID: 40327992 DOI: 10.1016/j.biopha.2025.118121] [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: 02/03/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Breast cancer stem cells (BCSCs) are a unique subpopulation of tumor cells driving tumor resistance, progression, metastasis, and recurrence. Reprogrammed cellular metabolism and key signaling pathways, including Wnt/β-catenin, TGF-β, STAT3, and PI3K/AKT/mTOR pathway play a vital role in maintaining BCSCs. Importantly, PI3K/Akt/mTOR pathway regulates metabolism, survival, growth, and invasion, with PIK3CA, encoding the PI3K catalytic subunit p110α, the most frequently mutated gene in breast cancer. This study investigates the effects of alpha-lipoic acid (LA) on the metabolic profile of BCSCs, focusing on its interaction with PI3K signaling. LA was found to bind PI3K, disrupting cancer-associated metabolic pathways and significantly inhibiting BCSC metabolism. Metabolomic analysis of MCF-7 and MDA-MB-231-derived breast cancer spheroids showed LA-induced metabolic shifts. In MCF-7 spheroids, LA induced upaccumulation of 15 metabolites and downaccumulation of 5, while in MDA-MB-231 spheroids, it induced upaccumulation of 3 and downaccumulation of 16. LA also enhanced the sensitivity of breast cancer spheroids to doxorubicin (Dox), demonstrating a synergistic effect. Mechanistically, LA modulates the PI3K/Akt/mTOR pathway, impairing cell survival and proliferation. These findings highlight the potential of LA as a therapeutic agent for reprogramming cancer metabolism and enhancing chemotherapy efficacy. These results provide a strong rationale for incorporating LA into combination therapy strategies for breast cancer treatment.
Collapse
Affiliation(s)
- Bandana Chakravarti
- Stem Cell/Cell Culture Lab, Center for Advanced Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh 226003, India.
| | - Manendra Singh Tomar
- Center for Advanced Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Faizan Abul Qais
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - K M Abdullah
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gunjan Sharma
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Archana Tewari
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Abhishek Yadav
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Pratima Gupta
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Center for Research in Anabolic Skeletal Target in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Ashutosh Shrivastava
- Center for Advanced Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Jawed Akhtar Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
15
|
Zaki RM, Ali MAM, Said M, Chaudhary AA, Boufahja F, Afzal O, Abu-Elsaoud AM, Abdel Halim AS. Molecular mechanisms underlying the effects of statins on bone metabolism: an evolving paradigm of statins delivery modalities for bone regeneration. Pharmacol Rep 2025; 77:624-644. [PMID: 40167878 DOI: 10.1007/s43440-025-00716-7] [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: 10/18/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025]
Abstract
Statins, recognized for their lipid-lowering capabilities, have demonstrated osteoanabolic and anti-resorptive effects on bone metabolism. The effects encompass the overexpression of bone morphogenetic proteins, heightened osteoblast activity, and the control of inflammation. Nevertheless, conventional systemic administration of statins has difficulties, including restricted bone bioavailability and possible adverse effects. Recent improvements in targeted and localized drug delivery are revolutionizing the therapeutic landscape for statins in bone applications. This review consolidates existing knowledge regarding the molecular processes by which statins influence bone metabolism and describes novel drug delivery methods such as nano-carriers, biomaterial scaffolds, and controlled-release systems. It seeks to address current knowledge deficiencies and offer insights into how enhanced bioavailability and specificity can optimize the efficiency of statins in bone regeneration. The review integrates molecular insights with novel pharmacological strategies to inform future research and clinical applications, pinpointing critical areas for exploration, such as optimal dose, delivery safety, and clinical efficacy.
Collapse
Affiliation(s)
- Randa Mohammed Zaki
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Mohamed A M Ali
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 11623, Saudi Arabia.
| | - Mayada Said
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 11623, Saudi Arabia
| | - Fehmi Boufahja
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 11623, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Abdelghafar M Abu-Elsaoud
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 11623, Saudi Arabia
| | - Alyaa S Abdel Halim
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| |
Collapse
|
16
|
He W, Hu X, Ge M, Meng K. The central role of ferroptosis-induced therapy mediated by tenacissoside H in anaplastic thyroid cancer. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119908. [PMID: 40311717 DOI: 10.1016/j.jep.2025.119908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/20/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tenacissoside H(TDH), a natural compound extracted from the dried vine stems of Marsdenia tenacissima (Roxb.) Wight et Arn., is considered to have anti-tumor effects. However, the anti-tumor activity of TDH against ATC remains unknown. AIM OF THE STUDY Ferroptosis, a novel form of programmed cell death, presents a promising target for therapeutic intervention, particularly in overcoming drug resistance in anaplastic thyroid carcinoma (ATC). We investigated the inhibitory effects of TDH on ATC cells, elucidating its ferroptosis-inducing mechanism, which to our knowledge has not been explored before. RESULTS Our findings indicate that TDH exerts an effect on the survival, proliferation, and migration of ATC cells. The strength of effect is dependent on dosage. Notably, ferroptosis marker proteins (GPX4, xCT, HO-1, TFR) were significantly downregulated following TDH treatment, whereas GPX4 and xCT expressions were partially restored post treatment with ferrostatin-1. Furthermore, in vivo studies confirmed that TDH effectively inhibited tumor growth in xenografted 8505C cells. CONCLUSIONS TDH could be considered a potential agent against ATC via inducing ferroptosis, providing a novel pharmacological basis for treating ATC.
Collapse
Affiliation(s)
- Wanwan He
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Xiaotian Hu
- Shanghai First People's Hospital, Shanghai, 200080, China
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China.
| | - Kexin Meng
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China.
| |
Collapse
|
17
|
Mantoan Ritter L, Annear NMP, Baple EL, Ben-Chaabane LY, Bodi I, Brosson L, Cadwgan JE, Coslett B, Crosby AH, Davies DM, Daykin N, Dedeurwaerdere S, Dühring Fenger C, Dunlop EA, Elmslie FV, Girodengo M, Hambleton S, Jansen AC, Johnson SR, Kearley KC, Kingswood JC, Laaniste L, Lachlan K, Latchford A, Madsen RR, Mansour S, Mihaylov SR, Muhammed L, Oliver C, Pepper T, Rawlins LE, Schim van der Loeff I, Siddiqui A, Takhar P, Tatton-Brown K, Tee AR, Tibarewal P, Tye C, Ultanir SK, Vanhaesebroeck B, Zare B, Pal DK, Bateman JM. mTOR pathway diseases: challenges and opportunities from bench to bedside and the mTOR node. Orphanet J Rare Dis 2025; 20:256. [PMID: 40426219 DOI: 10.1186/s13023-025-03740-1] [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: 11/06/2024] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a highly conserved serine/threonine kinase that regulates key cellular processes including cell growth, autophagy and metabolism. Hyperactivation of the mTOR pathway causes a group of rare and ultrarare genetic diseases. mTOR pathway diseases have diverse clinical manifestations that are managed by distinct medical disciplines but share a common underlying molecular basis. There is a now a deep understanding of the molecular underpinning that regulates the mTOR pathway but effective treatments for most mTOR pathway diseases are lacking. Translating scientific knowledge into clinical applications to benefit the unmet clinical needs of patients is a major challenge common to many rare diseases. In this article we expound how mTOR pathway diseases provide an opportunity to coordinate basic and translational disease research across the group, together with industry, medical research foundations, charities and patient groups, by pooling expertise and driving progress to benefit patients. We outline the germline and somatic mutations in the mTOR pathway that cause rare diseases and summarise the prevalence, genetic basis, clinical manifestations, pathophysiology and current treatments for each disease in this group. We describe the challenges and opportunities for progress in elucidating the underlying mechanisms, improving diagnosis and prognosis, as well as the development and approval of new therapies for mTOR pathway diseases. We illustrate the crucial role of patient public involvement and engagement in rare disease and mTOR pathway disease research. Finally, we explain how the mTOR Pathway Diseases node, part of the Research Disease Research UK Platform, will address these challenges to improve the understanding, diagnosis and treatment of mTOR pathway diseases.
Collapse
Affiliation(s)
- Laura Mantoan Ritter
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | - Nicholas M P Annear
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | - Leila Y Ben-Chaabane
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | - Istvan Bodi
- King's College Hospital NHS Foundation Trust, London, UK
| | | | | | | | | | | | | | | | | | | | - Frances V Elmslie
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | - Marie Girodengo
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
- The Francis Crick Institute, London, UK
| | - Sophie Hambleton
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Simon R Johnson
- Centre for Respiratory Research, NIHR Nottingham Biomedical Research Centre and Biodiscovery Institute, Translational Medical Sciences, University of Nottingham, Nottingham, UK
| | - Kelly C Kearley
- mTOR Node Advisory Panel (MAP), London, UK
- PTEN UK and Ireland Patient Group, London, UK
| | - John C Kingswood
- St George's University Hospitals NHS Foundation Trust, London, UK
| | | | - Katherine Lachlan
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Andrew Latchford
- Polyposis Registry, St Mark's Hospital, London, UK
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Sahar Mansour
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | | | | | - Tom Pepper
- PTEN Research, Cheltenham, Gloucestershire, UK
| | | | - Ina Schim van der Loeff
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ata Siddiqui
- King's College Hospital NHS Foundation Trust, London, UK
| | | | - Katrina Tatton-Brown
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | | | - Charlotte Tye
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | | | | | | | - Deb K Pal
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | - Joseph M Bateman
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK.
| |
Collapse
|
18
|
Xu W, Li X, He F, Zhao H, Wu J, Li M, Dai X, Li Y, Hu X, Li X, Cen J, Guo P, Duan S. Folate receptor-targeted pH-sensitive liposomes loaded with TGX-221 against prostate cancer by inhibiting PI3K/110β signaling. NANOSCALE ADVANCES 2025; 7:3267-3280. [PMID: 40212450 PMCID: PMC11979785 DOI: 10.1039/d5na00009b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/05/2025] [Indexed: 05/29/2025]
Abstract
Prostate cancer (PCa) is the most common cancer in men and the leading cause of cancer death worldwide. Overactivation of PI3K signaling has been reported to be associated with PCa. TGX221 is an effective specific inhibitor of PI3K, but its clinical application is greatly limited due to its poor solubility. Herein, by using folic acid-PEG-cholesterol semi-succinate (FA-PEG-CHEMS) as the targeting component, we developed a folate receptor-targeted pH-sensitive liposomal delivery system loaded with TGX221 (FA-Lip-TGX221) that could realize effective delivery and controlled release of drugs in the tumor. The prepared liposomes exhibited a uniform particle size and high stability. In addition, FA-Lip-TGX221 could be effectively internalized by PC-3 cells due to its ability to target folate receptors, thereby accumulating in tumor tissues. Meanwhile, in vitro and in vivo experiments suggested that FA-Lip-TGX221 could activate the PERK-ATF4-CHOP signaling pathway by inhibiting PI3K/110β signaling in PCa, thus significantly promoting endoplasmic reticulum (ER) stress-mediated cancer cell death. In conclusion, FA-Lip-TGX221 is a promising nano-delivery vehicle for the treatment of PCa, and also provide valuable references for all tumors overexpressing folate receptors.
Collapse
Affiliation(s)
- Weibo Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
- Medical School, Henan University Kaifeng 475004 China
| | - Xiaohan Li
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Fujin He
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Han Zhao
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Jing Wu
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Mengyu Li
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Xiaoying Dai
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Yanmin Li
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Xiaojiao Hu
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Xiaodong Li
- Medical School, Henan University Kaifeng 475004 China
| | - Juan Cen
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
- The First Affiliated Hospital of Henan University Kaifeng Henan 475004 PR China
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
| | - Shaofeng Duan
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
- The First Affiliated Hospital of Henan University Kaifeng Henan 475004 PR China
| |
Collapse
|
19
|
Chen Y, Xu R, Xie B, Ma L, He Y, Liu H, Chen T. Ultrasound-Driven Selenium Nanoparticles Realize Bone Defect Repair through Activating Selenoproteins to Regulate PI3K/AKT Signaling Pathway. ACS NANO 2025; 19:18256-18269. [PMID: 40338671 DOI: 10.1021/acsnano.4c18240] [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: 05/10/2025]
Abstract
Excessive and variable inflammation in bone defects is a key factor that impedes effective bone repair. Herein, an ultrasound-controlled composite hydrogel (LNT-SeNPs@Gel) integrating gelatin-methacryloyl and lentinan-decorated selenium nanoparticles (LNT-SeNPs) is developed, exhibiting strong antioxidant and anti-inflammatory properties to remodel the inflammatory microenvironment of bone defects. This hydrogel serves as a platform for integrating bifunctional ultrasound (ultrasound modulation, USc and ultrasound for repairing, USr), facilitating cascade treatment and reducing the overall treatment period. During the inflammatory phase of bone repair, USc remotely modulates the LNT-SeNPs@Gel hydrogel, regulating the release of LNT-SeNPs to inhibit the overproduction of reactive oxygen species (ROS) and inflammatory factors, ultimately remodeling the inflammatory microenvironment. Subsequently, USr could activate the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling pathway regulated by selenoproteins to enhance the osteogenesis of MC3T3-E1 cells, thereby accelerating the bone repair process. Consequently, the combination of bifunctional ultrasound and LNT-SeNPs@Gel significantly improves bone repair outcomes and reduces the treatment period in rats. In conclusion, this study implies that the coordinated integration of the dual effects of ultrasound is a promising strategy for handling the complex and lengthy bone defects repair.
Collapse
Affiliation(s)
- Yufan Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Renhao Xu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Bin Xie
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Li Ma
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Yanni He
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Hongmei Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- Guangdong Engineering Technology Research Center of Emergency Medicine, Guangzhou 510317, China
| | - Tianfeng Chen
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| |
Collapse
|
20
|
Zhong L, Purushothaman B, Tu Q, Boopathi S, Zhang Y. GCNT3-mediated glycosylation in cancer biology: Implications for tumorigenesis, metastasis, and therapeutic targeting. Int J Biol Macromol 2025; 315:144427. [PMID: 40403799 DOI: 10.1016/j.ijbiomac.2025.144427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 05/16/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
Glycosylation is a fundamental post-translational modification that plays a pivotal role in cancer progression, influencing cell adhesion, immune evasion, metastasis, and drug resistance. Among glycosyltransferases, Core 2 β-1,6-N-acetylglucosaminyltransferase 3 (GCNT3) has emerged as a key regulator of tumor behavior, with its effects varying across different cancers. While elevated GCNT3 expression is associated with better prognosis and chemotherapy response in ovarian cancer, it correlates with poor survival, tumor invasiveness, and immune suppression in pancreatic and lung cancers. This dual nature underscores the complexity of GCNT3's role in cancer biology. As a biomarker, GCNT3 has shown potential for prognostic and therapeutic applications, particularly in colorectal and ovarian cancers. Targeting GCNT3 therapeutically presents challenges due to its role in normal physiological glycosylation, and the lack of selective inhibitors. Current research suggests that GCNT3-targeted therapies, in combination with immunotherapy or chemotherapy, could improve treatment outcomes by modulating mucin production, tumor metabolism, and immune responses. This review critically explores GCNT3's diverse functions, its impact on cancer progression, and its potential as a therapeutic target, highlighting the need for cancer-specific approaches and future innovations in drug development to harness its clinical potential effectively.
Collapse
Affiliation(s)
- Lin Zhong
- Institute of Synthetic Biology Industry, Hunan University of Arts and Science, Changde 415000, China
| | | | - Qiang Tu
- Institute of Synthetic Biology Industry, Hunan University of Arts and Science, Changde 415000, China; Helmholtz International Lab for Anti-Infectives, Shandong University-Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| | - Seenivasan Boopathi
- Helmholtz International Lab for Anti-Infectives, Shandong University-Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China.
| | - Youming Zhang
- Institute of Synthetic Biology Industry, Hunan University of Arts and Science, Changde 415000, China; Helmholtz International Lab for Anti-Infectives, Shandong University-Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China; Shenzhen Key Laboratory of Genome Manipulation and Biosynthesis, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
21
|
Erdman V, Petintseva A, Timasheva Y, Tuktarova I, Nasibullin T, Korytina G. Genetic predictors of longevity and survival in cellular homeostasis genes: A case-control study. Gene 2025; 962:149576. [PMID: 40398646 DOI: 10.1016/j.gene.2025.149576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 05/05/2025] [Accepted: 05/17/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Longevity is defined by the ability to maintain both physical and mental health throughout a long life and may results from adaptive mechanisms that mitigate aging's detrimental effects. METHODS The 20-year follow-up study of 3,312 unrelated individuals aged 18-114 years from the Volga-Ural region of Eurasia investigated variants in cellular homeostasis genes IGF1, PIK3R1, AKT1, MTOR, NFE2L2, KEAP1, HIF1A, TP53, and SIRT1, to identify associations with clinical aging phenotypes and healthy longevity. RESULTS In men, KEAP1 (rs1048290) CC genotype was a longevity and survival marker (OR = 2.39, P = 3E-05, HR = 0.54, P = 2.4E-03). NFE2L2 (rs6721961) TT genotype was linked to higher mortality (HR = 1.77, P = 0.031), particularly combined with KEAP1 (rs1048290) G and AKT1 (rs3803304) C alleles (HR = 2.8, P = 0.023). In women, AKT1 (rs3803304) C allele interacted with NFE2L2 (rs6721961) TT genotype (SF = 0.13, P = 3.6E-03), and was linked to longevity (OR = 2.22, P = 6.3E-03) and protection against cerebrovascular diseases (OR = 0.62, P = 5.1E-03). AKT1 (rs3803304) GG genotype, along with HIF1A (rs11549465) T and SIRT1 (rs3758391) T alleles (SF = 2.52, P = 1.5E-03), promoted survival (HR = 0.71, P = 0.014). In men, HIF1A (rs11549465) TT genotype predicted cardiovascular mortality (HR = 7.5, P = 5.5E-03). SIRT1 (rs3758391) TT genotype was associated with improved survival in individuals with diabetes (HR = 0.4, P = 5.8E-03) and multimorbidity (HR = 0.48, P = 0.025). CONCLUSION Variants in NFE2L2, KEAP1, SIRT1, AKT1, and HIF1A, along with their interactions, were significantly associated with survival in age-related diseases and healthy longevity.
Collapse
Affiliation(s)
- Vera Erdman
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia; Bashkir State Medical University, Ufa 450008, Russia.
| | - Anna Petintseva
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia
| | - Yanina Timasheva
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia; Bashkir State Medical University, Ufa 450008, Russia
| | - Ilsiar Tuktarova
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia
| | - Timur Nasibullin
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia
| | - Gulnaz Korytina
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia; Bashkir State Medical University, Ufa 450008, Russia
| |
Collapse
|
22
|
Martinez-Mendoza F, Andonegui-Elguera S, Sosa-Eroza E, Gomez-Apo E, Escobar-España A, Gonzalez-Torres C, Gaytan-Cervantes J, Palma-Guzman A, Torres-Flores H, Moscona-Nissan A, Hinojosa-Alvarez S, Hernandez-Perez J, Chavez-Santoscoy RA, Guinto G, Guinto-Nishimura GY, Lopez-Felix BE, Zepeda-Fernandez EU, Estrada-Estrada EM, Correa-Correa V, Gonzalez-Zavala PA, Asenscio-Montiel MA, Garcia-Vargas MA, Cantu-Chavez E, Arreola-Rosales RL, Taniguchi-Ponciano K, Marrero-Rodriguez D, Mercado M. Spatial transcriptomics reveal PI3K-AKT and metabolic alterations in aggressive, treatment-resistant lactotroph pituitary neuroendocrine tumors. Acta Neuropathol Commun 2025; 13:107. [PMID: 40390063 PMCID: PMC12087103 DOI: 10.1186/s40478-025-02025-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/30/2025] [Indexed: 05/21/2025] Open
Abstract
Clinically aggressive lactotroph pituitary neuroendocrine tumors (PitNET) are invasive tumors with an unusually rapid growth rate despite maximally tolerated doses of dopamine agonist (DA). We aimed to unravel the molecular heterogeneity of lactotroph PitNET and to identify biomarkers of aggressiveness and resistance to pharmacological treatment. A total of 13 patients harboring DA-resistant lactotroph PitNET were included in this study. Visium Spatial Transcriptomics (ST), whole transcriptome sequencing (WTS), and whole exome sequencing (WES) were performed in tumors from 4 of these patients; WTS and WES was carried out in 5; tumors from two patients underwent ST and WES and tumors from two other patients underwent only ST. Tumors were classified as null or partial responders according to their response to DA treatment. The eight PitNET analyzed by ST exhibited significant intratumoral heterogeneity, with clones showing alterations in PI3K/AKT and lipid metabolism pathways, particularly inositol phosphate, glycerophospholipid, and sphingolipid metabolism. The cell-cell communication analysis showed FGF-FGFR ligand receptor interaction whilst the transcription factors RXRA and CREM showed participation in both groups. A trajectory exploration was performed by including all PitNET together in a single analysis to determine whether there was a tendency or molecular pathway showing a differentiation pattern that would guide the transition from a partially responsive PitNET to a completely unresponsive one. We did not observe any such pattern. All of these findings were corroborated in the cohort of DA-resistant PitNETs in which only bulk WTS and WES were performed. The bulk WTS corroborated lipid metabolism and PI3K-AKT pathway alteration in PitNET, whereas the WES showed only SF3β1 and TP53 variants in one tumor each. Our work suggests that the PI3K/AKT pathway may constitute a molecular target at which to aim therapeutic strategies designed to treat aggressive and DA-resistant lactotroph PitNET.
Collapse
Affiliation(s)
- Florencia Martinez-Mendoza
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Sergio Andonegui-Elguera
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Ernesto Sosa-Eroza
- Servicio de Endocrinología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Erick Gomez-Apo
- Área de Neuropatología, Servicio de Anatomía Patológica, Hospital General de México Dr. Eduardo Liceaga, Ciudad de México, México
| | - Aurea Escobar-España
- Área de Neuropatología, Servicio de Anatomía Patológica, Hospital General de México Dr. Eduardo Liceaga, Ciudad de México, México
| | - Carolina Gonzalez-Torres
- Laboratorio de Secuenciación, División de Desarrollo de la Investigación, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Javier Gaytan-Cervantes
- Laboratorio de Secuenciación, División de Desarrollo de la Investigación, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Alam Palma-Guzman
- Laboratorio de Histología, Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Hugo Torres-Flores
- Servicio de Anatomía Patológica, Hospital General de México Dr. Eduardo Liceaga, Ciudad de México, México
| | - Alberto Moscona-Nissan
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | | | | | | | - Gerardo Guinto
- Centro Neurológico, Centro Médico ABC, Ciudad de México, México
| | - Gerardo Y Guinto-Nishimura
- Departamento de Neurocirugia, Intituto Nacional de Neurologia y Neurocirugia "Manuel Velasco Suarez", Ciudad de Mexico, Mexico
| | - Blas E Lopez-Felix
- Servicio de Neurocirugia, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Erick U Zepeda-Fernandez
- Servicio de Neurocirugia, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Erick M Estrada-Estrada
- Servicio de Neurocirugia, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Victor Correa-Correa
- Servicio de Neurocirugia, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Pedro A Gonzalez-Zavala
- Servicio de Neurocirugia, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Marco A Asenscio-Montiel
- Servicio de Neurocirugia, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Miguel A Garcia-Vargas
- Servicio de Neurocirugia, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Emmanuel Cantu-Chavez
- Servicio de Neurocirugia, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Rocio L Arreola-Rosales
- Servicio de Patologia, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Keiko Taniguchi-Ponciano
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México.
| | - Daniel Marrero-Rodriguez
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México.
| | - Moisés Mercado
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México.
| |
Collapse
|
23
|
Stachyra P, Grzybowska-Szatkowska L. Signaling Pathways in Gliomas. Genes (Basel) 2025; 16:600. [PMID: 40428422 DOI: 10.3390/genes16050600] [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: 03/30/2025] [Revised: 05/10/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Changes in cell signaling pathways, which in normal conditions determine the maintenance of cell homeostasis and the correctness of its basic processes, may cause the transformation of a normal cell into a cancer cell. Alterations in cellular metabolism leading to oncogenesis are considered to be a hallmark of cancer cells. Therefore, a thorough understanding of cellular enzymes affecting metabolism and respiration, as well as intracellular pathways connected with them, seems crucial. These changes may be both prognostic and predictive factors, especially in terms of using molecularly targeted therapies. Aberrations in the pathways responsible for cell growth and angiogenesis are considered particularly important in the process of oncogenesis. Gliomas are the most common primary malignant tumors of the brain. The most important molecular disorders determining their particularly malignant nature are aberrations in the pathways responsible for cell growth and angiogenesis, such as the PI3K/Akt or RAS/MAPK/ERK signaling pathway, as well as excessive activity of enzymes, like hexokinases, which play a key role in glycolysis, autophagy, and apoptosis. The multitude of alterations detected in glioma cells, high heterogeneity, and the immunosuppressive environment within the tumor are the main features causing failures in the attempts to implement modern therapies.
Collapse
Affiliation(s)
- Paulina Stachyra
- II Department of Oncology and Clinical Immunology with Day Chemotherapy, Oncology Centre of the Lublin Region, Jaczewskiego 7, 20-090 Lublin, Poland
- Department of Radiotherapy, Medical University of Lublin, Chodźki 7, 20-093 Lublin, Poland
| | | |
Collapse
|
24
|
Wang H, Zhu J, Wang H, Zheng W, Wang L, Zhu J, Wang Z, Du Q. The role of FAM111B in the malignant progression and molecular regulation of human glioma through the PI3K/Akt pathway. Chin Neurosurg J 2025; 11:9. [PMID: 40390043 PMCID: PMC12087166 DOI: 10.1186/s41016-025-00395-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 04/14/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Gliomas represent the most prevalent primary neoplasm in the adult central nervous system. Despite advancements in therapeutic modalities, such as surgical intervention, radiotherapy, chemotherapy, and tumor treatment, the 5-year survival rate of glioma patients remains low. Therefore, there is an urgent need to develop additional treatment methods. Recent studies have suggested that FAM111B is involved in DNA repair, cell cycle regulation, and apoptosis. FAM111B mutations and overexpression are related to cancer. METHODS We found that FAM111B was significantly overexpressed in glioma tissues compared to the adjacent tissues by analyzing data from the TCGA_GBM&LGG and CGGA databases. Moreover, overexpression of FAM111B was associated with shorter overall survival, and disease-specific survival and tended to increase with disease stage progression. Cellular experiments confirmed these results. These results suggest that overexpression of FAM111B promotes the proliferation, migration, and invasion of glioma cells, whereas the knockdown of FAM111B inhibits these activities. We also found that FAM111B regulated glioma cell proliferation, migration, and invasion via the PI3K/AKT pathway. RESULTS FAM111B is capable of enhancing the proliferation, invasion, and migration capabilities of glioma cells and promotes the malignant progression of glioma via the PI3K/Akt signaling pathway. CONCLUSIONS This is the first study to demonstrate that FAM111B plays a crucial role in the proliferation, migration, and invasion of glioma cells. The malignant phenotype of FAM111B has also been shown to be closely associated with the PI3K/AKT pathway. FAM111B may be a predictive biomarker and a potential therapeutic target for gliomas.
Collapse
Affiliation(s)
- Heng Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Junrou Zhu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Haiyang Wang
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Wenhao Zheng
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Linjie Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jinhao Zhu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Zheng Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| |
Collapse
|
25
|
Zhang X, Kang H, Li B, Xiong Y, Zheng S, Zhang D, Liu Y, Li S, Liu Y, Liu H, Gao Y, Ma L. Structural Optimization of 1,3-Diaryl-1,2,4-triazole-Capped Histone Deacetylase 6 Inhibitors to Obtain Novel Antiesophageal Cancer Candidates. J Med Chem 2025. [PMID: 40382720 DOI: 10.1021/acs.jmedchem.4c03231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Esophageal cancer, a leading global cancer, lacks effective therapies. Inhibition of histone deacetylase 6 (HDAC6) is a promising antitumor strategy, yet its role in esophageal cancer remains underexplored. Through structural optimization of our previously developed 1,3-diaryl-1,2,4-triazole-capped HDAC6 inhibitors, we identified compound 38k, exhibiting remarkably enhanced HDAC6 inhibition (IC50 = 3.12 nM) and 352-fold selectivity over HDAC1. Molecular docking analysis, CETSA, and BLI confirmed its strong HDAC6 binding. Moreover, 38k displayed robust in vitro and in vivo antiesophageal cancer efficacy, along with an advantageous pharmacokinetic and safety profile. Notably, combining 38k with a PI3K inhibitor synergistically enhanced the efficacy (75.02% tumor growth inhibition vs 50.94% monotherapy), likely by counteracting HDAC6 inhibition-induced PI3K/AKT activation. These findings validate HDAC6 as a therapeutic target and highlight 38k as a promising candidate for esophageal cancer treatment, particularly in combination regimens.
Collapse
Affiliation(s)
- Xinhui Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
- Newland Pharmaceutical Co., Ltd., Xuchang, Henan 461500, China
| | - Huiqin Kang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Bingqian Li
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yuhan Xiong
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Shuxian Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Di Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yuanfan Liu
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Shiyu Li
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Ying Liu
- The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou, Henan 450001, China
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Liying Ma
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
- Key Laboratory of Cardio-cerebrovascular Drug, China Meheco Topfond Pharmaceutical Co., Zhumadian 463000, China
| |
Collapse
|
26
|
Zhu T, Wang P, Wang R, Tong G, Sun Y, Yang S, Zhou X, Mou D, Zhong W, Cai Y. Propranolol accelerates adipogenesis and inhibits endothelium differentiation of HemSCs via suppressing HK2 mediated glycolysis. Pediatr Res 2025:10.1038/s41390-025-04080-3. [PMID: 40382470 DOI: 10.1038/s41390-025-04080-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/27/2025] [Accepted: 04/13/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND As the first-line treatment for hemangioma (IH), the mechanism of propranolol (PRN) remains unclear. In clinical practice, challenges such as PRN resistance and rebound after discontinuation of PRN are frequently encountered. Hence, this research seeks to investigate the mechanisms underlying PRN-induced regression of IH. METHODS Hexokinase 2 (HK2) expression was assessed via immunohistochemistry and double-labeling staining. Glycolysis in hemangioma-derived stem cells (HemSCs) was evaluated by measuring glucose uptake, lactate, and ATP production. Peroxisome proliferator-activated receptor Gamma (PPARγ) and vascular endothelial cadherin (VE-cadherin) levels were analyzed using Western blot and qPCR. PRN-treated HemSCs were examined for adipogenic differentiation via Oil Red O and BODIPY staining. RESULTS Our results demonstrate that PRN inhibits HemSCs proliferation and endothelial differentiation while promoting adipogenesis by suppressing glycolysis. This effect occurs through HK2 downregulation, likely mediated by PI3K-Akt pathway inhibition. Notably, HK2 expression was significantly lower in CD133+ cells from involutive hemangiomas versus proliferative lesions. CONCLUSIONS This study presents the first evidence for the essential role of glycolysis in regulating the proliferation and differentiation of HemSCs, while the efficiency of PRN may be associated with the inhibition of HK2-mediated glycolysis in HemSCs by suppressing the activities of PI3K-Akt pathway. IMPACT Glycolysis level is high in HemSCs. Glycolysis is required in propranolol perturbated the HemSCs differentiation. PRN could inhibit glycolysis of HemSCs through down-regulation of HK2 expression. PRN suppressed endothelial differentiation and accelerated adipogenesis of HemSCs. PRN down-regulated HK2 expression through restrained the PI3k-Akt pathway. Schematic of treatment mechanism of PRN in IH. HK2 is highly expressed in HemSCs. PRN may be associated with the inhibition of HK2-mediated glycolysis in HemSCs by suppressing the activities of PI3K-Akt pathway which finally promotes regression of IH.
Collapse
Affiliation(s)
- Tianshuang Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral & Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Peipei Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral & Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Rong Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guoyong Tong
- Department of Stomatology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Yu Sun
- Department of Plastic Surgery, Wuhan Children's Hospital, Wuhan, China
| | - Shaodong Yang
- Department of Pathology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | | | | | - Wenqun Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral & Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Yu Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral & Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
27
|
Liu H, Zhou C, Yan Z, Yang H, Zhao Y, Tian R, Bo X, Zhao L, Ren W. Sialyltransferase gene signature as a predictor of prognosis and therapeutic response in kidney renal clear cell carcinoma. Discov Oncol 2025; 16:785. [PMID: 40377806 PMCID: PMC12084485 DOI: 10.1007/s12672-025-02566-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 05/05/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Sialyltransferases are enzymes involved in the addition of sialic acid to glycoproteins and glycolipids, influencing various physiological and pathological processes. The expression and function of sialyltransferases in tumors, particularly in kidney renal clear cell carcinoma (KIRC) remained underexplored. This study aimed to develop a prognostic model based on sialyltransferase-related genes (SRGs) to predict the prognosis and treatment response of patients with KIRC. METHODS We utilized RNA-Seq data of KIRC from The Cancer Genome Atlas (TCGA) database, selecting samples with survival data and clinical outcomes. Somatic mutation and neoantigen data were analyzed using the "maftools" package, and genes involved in the sialylation process were identified through the Molecular Signatures Database. Validation cohorts of KIRC samples were obtained from the International Cancer Genome Consortium (ICGC) database. Single-cell RNA sequencing (scRNA-seq) data were downloaded from the Gene Expression Omnibus (GEO) platform, and preprocessing, normalization, and dimensionality reduction analyses were conducted using the "Seurat" package. Differentially expressed sialylation genes were identified using the "limma" package, and their functional enrichment was assessed via Gene Ontology GO and KEGG analyses. Consensus clustering analysis was performed to identify molecular subtypes of KIRC based on sialylation, and drug sensitivity of different subtypes was evaluated using the "pRRophetic" package. A risk signature model comprising 5 SRGs was constructed through univariate and multivariate Cox regression analyses and validated in both the TCGA and ICGC cohorts. The "estimate" package was utilized to calculate immune and stromal scores for each KIRC sample, assessing the tumor immune microenvironment characteristics of different subtypes. RESULTS Analysis of scRNA-seq data identified 25 cell subtypes, categorized into 9 cell types. CD4 + memory cells exhibited the highest potential interactions with other cell subtypes. We identified 14 differentially expressed sialylation genes and confirmed their enrichment in various biological pathways through GO and KEGG analyses. Consensus clustering analysis based on sialylation identified 2 molecular subtypes: C1 and C2. The C2 subtype demonstrated higher sialylation scores and poorer prognosis. Drug sensitivity analysis indicated that the C1 subtype had better responses to Dasatinib and Lapatinib, whereas the C2 subtype was more sensitive to Epothilone B and Vinorelbine. The risk signature model, constructed with five distinct SRGs, exhibited strong predictive accuracy, as indicated by Area Under the Curve (AUC) values of 0.68, 0.69, and 0.70 for 1-, 3-, and 5-year survival, respectively, across both the TCGA and ICGC validation cohorts. Immune microenvironment analysis revealed that the C1 subtype exhibited higher immune and stromal scores, while the C2 subtype showed significantly enhanced expression of immune checkpoint genes. CONCLUSION This study successfully developed a prognostic model based on SRGs, effectively predicting the prognosis and drug response of KIRC patients. The model demonstrated significant predictive performance and potential clinical application value. Furthermore, the study highlighted the critical role of sialylation in KIRC, offering new insights into its underlying mechanisms in tumor biology. These findings could guide personalized treatment strategies for KIRC patients, emphasizing the importance of sialylation in cancer prognosis and therapy.
Collapse
Affiliation(s)
- Huiyu Liu
- Department of Urology, Dongying People's Hospital, Dongying, 257091, Shandong, China.
| | - Changwei Zhou
- Department of Urology, Dongying People's Hospital, Dongying, 257091, Shandong, China
| | - Zaichun Yan
- Department of Urology, Dongying People's Hospital, Dongying, 257091, Shandong, China
| | - Hairong Yang
- Department of Critical Care Medicine, Dongying People's Hospital, Dongying, 257091, Shandong, China
| | - Yun Zhao
- Department of Urology, Dongying People's Hospital, Dongying, 257091, Shandong, China
| | - Rui Tian
- Dongying District Traditional Chinese Medicine Hospital, Dongying, 257029, Shandong, China
| | - Xuejun Bo
- Department of Urology, Dongying People's Hospital, Dongying, 257091, Shandong, China
| | - Leizuo Zhao
- Department of Urology, Dongying People's Hospital, Dongying, 257091, Shandong, China
| | - Wei Ren
- Department of Urology, Dongying People's Hospital, Dongying, 257091, Shandong, China
| |
Collapse
|
28
|
Zhao C, Kong K, Liu P, Chen X, Rong K, Zhang P, Wang L, Wang X. Regulating obesity-induced osteoarthritis by targeting p53-FOXO3, osteoclast ferroptosis, and mesenchymal stem cell adipogenesis. Nat Commun 2025; 16:4532. [PMID: 40374649 PMCID: PMC12081733 DOI: 10.1038/s41467-025-59883-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/08/2025] [Indexed: 05/17/2025] Open
Abstract
Obesity-related osteoarthritis (OA) and the molecular mechanisms governing multiple joint structural changes that occur with obesity are not well understood. This study investigated the progression of obesity in mice and validated the results using human joint samples post-arthroplasty. The results show that obesity is associated with the degeneration of the cartilage layer and abnormal remodeling of the subchondral bone layer, and this occurs alongside aging and DNA damage in chondrocytes, osteoclasts, and stem cells. Regulation of p53-FOXO3 gene loop expression in response to DNA damage effectively inhibits chondrocyte apoptosis, catabolism, and excessive osteoclast differentiation, while the intra-articular delivery of a lentivirus expressing FOXO3 to mouse joints alleviates the progression of OA. The excessive differentiation of subchondral bone marrow osteoclasts is ferroptosis-dependent and driven by the senescence-associated secretory phenotype. The results have identified multiple potential targets for future research into the progression of obesity-related OA.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keyu Kong
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengcheng Liu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuzhuo Chen
- Department of Oral Surgery, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kewei Rong
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pu Zhang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaoqing Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
29
|
Kobayashi K, Yasui Y, Uehara T, Ninomiya K, Miwa H, Han L. Nicotinic acetylcholine receptors modulate casein secretion and claudin expression in mammary epithelial cells by regulating STAT5 and Akt pathways. Biochem Biophys Res Commun 2025; 770:152036. [PMID: 40382844 DOI: 10.1016/j.bbrc.2025.152036] [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: 05/13/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Smoking induces a decrease in breast milk volume, adverse changes in milk composition, and a shorter lactation period in breastfeeding women. In breastfeeding women, nicotine from tobacco is transferred from the blood to breast milk. Previously, we reported that nicotine adversely affects milk production and tight junctions (TJs) in mammary epithelial cells (MECs) in vitro. However, the mechanisms by which nicotine influences milk production and TJs in MECs remain unclear. During lactation, MECs are in contact with acetylcholine (ACh) in milk and express multiple nicotinic ACh receptors (nAChRs). In this study, we investigated whether nAChRs and ACh are involved in milk production TJs in MECs using a culture model of MECs that exhibit milk production ability and formation of less-permeable TJs. The results showed that nAChRα2 and nAChRα3 agonists, Br-PBTC and NS3861, respectively, suppressed casein secretion and increased claudin-4, a TJ protein. In addition, Br-PBTC and NS3861 inactivated STAT5 and Akt, which are signaling molecules that facilitate milk production in MECs. However, ACh did not influence casein secretion, claudin expression, or the activation of STAT5 and Akt in MECs. In contrast, the acetylcholinesterase inhibitor (donepezil) and nAChRα3 antagonist (α-conotoxin PIA) inhibited casein secretion concurrently inactivating STAT5 and Akt. Furthermore, short-term treatment with Br-PDTC and NS3861 on the apical side of MECs induced the inactivation of STAT5 and Akt. These findings indicate that MECs regulate milk production and TJ formation by regulating the acetylcholine levels in milk and that nicotine adversely affect milk production in MEC by disrupting the ACh/nAChR axis.
Collapse
Affiliation(s)
- Ken Kobayashi
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan.
| | - Yuki Yasui
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| | - Tamaki Uehara
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| | - Kazuki Ninomiya
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| | - Hideki Miwa
- Molecular Neuropsychopharmacology Section, Department of Neuropsychopharmacology, National Institute of Mental Health: National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, 187-8553, Tokyo, Japan
| | - Liang Han
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| |
Collapse
|
30
|
Pinto M, Violante S, Cascão R, Faria CC. Unlocking the Role of Metabolic Pathways in Brain Metastatic Disease. Cells 2025; 14:707. [PMID: 40422210 DOI: 10.3390/cells14100707] [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: 03/18/2025] [Revised: 04/30/2025] [Accepted: 05/09/2025] [Indexed: 05/28/2025] Open
Abstract
The dissemination of malignant cells to the brain is a late-stage complication of cancer, leading to significant morbidity and mortality. Brain metastases (BM) affect 20-30% of cancer patients, primarily originating from lung cancer, breast cancer, and melanoma. Despite advances in molecular-targeted therapies, brain metastatic disease remains incurable, with a poor median survival of ≤12 months if left untreated. The lack of therapeutic efficacy is mainly attributed to the presence of the blood-brain barrier (BBB) and genetic differences between BM and their primary tumors. Previously published data have identified potential driver mutations of BM. However, the mechanisms underlying brain cancer dissemination remain unknown. Recent studies emphasize the pivotal role of metabolic adaptations in supporting the metastatic process, particularly in the nutrient-poor microenvironment characteristic of the brain. Understanding the interplay between metabolism and genetic alterations associated with brain metastatic disease could unveil novel therapeutic targets that are more effective in treating patients. This review focuses on relevant metabolic pathways in cancer, particularly brain cancer dissemination, while also presenting information on current preclinical models of BM, relevant clinical trials, and preclinical studies targeting metabolic reprogramming, providing an overview for advancing therapeutic strategies in BM.
Collapse
Affiliation(s)
- Madalena Pinto
- GIMM-Gulbenkian Institute for Molecular Medicine, Avenida Prof. Egas Moniz, 1649-035 Lisboa, Portugal
| | - Sara Violante
- GIMM-Gulbenkian Institute for Molecular Medicine, Avenida Prof. Egas Moniz, 1649-035 Lisboa, Portugal
| | - Rita Cascão
- GIMM-Gulbenkian Institute for Molecular Medicine, Avenida Prof. Egas Moniz, 1649-035 Lisboa, Portugal
| | - Claudia C Faria
- GIMM-Gulbenkian Institute for Molecular Medicine, Avenida Prof. Egas Moniz, 1649-035 Lisboa, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Unidade Local de Saúde de Santa Maria (ULSSM), Avenida Prof. Egas Moniz, 1649-035 Lisboa, Portugal
- Clínica Universitária de Neurocirurgia, Faculdade de Medicina da Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-035 Lisboa, Portugal
| |
Collapse
|
31
|
Zeng Y, Tao Y, Du G, Huang T, Chen S, Fan L, Zhang N. Advances in the mechanisms of HIF-1α-enhanced tumor glycolysis and its relation to dedifferentiation. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 197:1-10. [PMID: 40373959 DOI: 10.1016/j.pbiomolbio.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Metabolic reprogramming, a hallmark of malignancy, enables tumor cells to adapt to the harsh and dynamic tumor microenvironment (TME) by altering metabolic pathways. Hypoxia, prevalent in solid tumors, activates hypoxia inducible factor 1α (HIF-1α). HIF-1α drives metabolic reprogramming, enhancing glycolysis primarily through the Warburg effect to reduce oxygen dependence and facilitate tumor cell growth/proliferation. The above process is associated with accelerated tumor cell dedifferentiation and enhanced stemness, generating cancer stem cells (CSCs) which possesses the potential for self-renewal and differentiation that can differentiate into a wide range of subtypes of tumor cells and fuel tumor heterogeneity, metastasis, and recurrence, complicating therapy. This review examines the HIF-1α-glycolysis-dedifferentiation crosstalk mechanisms, expecting that indirect inhibition of HIF-1α by targeting metabolic enzymes, metabolites, or their signaling pathways will offer an effective therapeutic strategy to improve the cancer treatment outcomes.
Collapse
Affiliation(s)
- Yu Zeng
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yonggang Tao
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Guotu Du
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tianyu Huang
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shicheng Chen
- Department of Urology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Longmei Fan
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Neng Zhang
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
32
|
Wu Y, Wang B, Mao X, Chen W, Akber Aisa H. Harmine derivative B-9-3 inhibits non-small cell lung cancer via the VEGFA/PI3K/AKT pathway. Front Pharmacol 2025; 16:1526952. [PMID: 40432889 PMCID: PMC12107193 DOI: 10.3389/fphar.2025.1526952] [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: 11/12/2024] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Background This study aimed to investigate the molecular mechanism by which the Harmine derivative B-9-3 inhibits angiogenesis and promotes apoptosis in non-small cell lung cancer (NSCLC). Methods Three non-small cell lung cancer (NSCLC) models (human NSCLC cell line A549, human lung squamous cell carcinoma cell line H226, human large cell lung carcinoma cell line H460) were established. Cell proliferation was assessed using CCK-8 assays and colony formation assays. Cell motility was evaluated through scratch wound healing, invasion, and migration assays. Cell apoptosis was analyzed by Hoechst 33258 staining, AO/EB fluorescence staining, and flow cytometry. Real-time PCR was used to measure the mRNA expression of B-cell lymphoma/leukemia-2 (Bcl-2), Bcl-2-associated X protein (Bax), and Caspase-3, while Western blotting was performed to assess the protein levels of vascular endothelial growth factor A (VEGFA), phosphatidylinositol 3-kinases p110 Beta (PI3K), phospho-phosphatidylinositol 3-kinases (p-PI3K), protein kinase B (AKT), phosphorylated protein kinase B (p-AKT), Bax, Bcl-2, and Caspase-3. Results Compared to the control group, B-9-3 (50, 100, 200 μg/mL) inhibited the growth and motility of the three types of lung cancer cells, suppressed cell invasion and migration, and promoted cell apoptosis and necrosis. The apoptosis rates in three types of non-small cell lung cancer (NSCLC) cells were significantly increased. The mRNA expressions of Bax and Caspase-3 were markedly upregulated, while that of Bcl-2 was significantly downregulated. Additionally, the protein levels of VEGFA, p-PI3K/PI3K, p-AKT/AKT, and Bcl-2 were notably reduced, whereas the protein levels of Bax and Caspase-3 were significantly elevated. Conclusion The harmine derivative B-9-3 may exert its anti-NSCLC effects by inhibiting angiogenesis and promoting lung cancer cell apoptosis via the VEGFA/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yuche Wu
- Xinjiang Technical Institute of Physics and Chemistry Chinese Academy of Sciences, Urumqi, Xinjiang, China
| | - Bing Wang
- The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xuwen Mao
- College of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Wei Chen
- Xinjiang Huashidan Pharmaceutical Co., Ltd., Urumqi, Xinjiang, China
| | - Haji Akber Aisa
- Xinjiang Technical Institute of Physics and Chemistry Chinese Academy of Sciences, Urumqi, Xinjiang, China
| |
Collapse
|
33
|
Zeng C, Gao Y, Lan B, Wang J, Ma F. Metabolic reprogramming in cancer therapy-related cardiovascular toxicity: Mechanisms and intervention strategies. Semin Cancer Biol 2025; 113:39-58. [PMID: 40349808 DOI: 10.1016/j.semcancer.2025.05.009] [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: 01/07/2025] [Revised: 04/20/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Cancer therapy-related cardiovascular toxicity (CTR-CVT) poses a major challenge in managing cancer patients, contributing significantly to morbidity and mortality among survivors. CTR-CVT includes various cardiovascular issues, such as cardiomyopathy, myocardial ischemia, arrhythmias, and vascular dysfunction, which significantly impact patient prognosis and quality of life. Metabolic reprogramming, characterized by disruptions in glucose, lipid, and amino acid metabolism, represents a shared pathophysiological feature of cancer and cardiovascular diseases; however, the precise mechanisms underlying CTR-CVT remain inadequately understood. In recent years, strategies targeting metabolic pathways have shown promise in reducing cardiovascular risks while optimizing cancer treatment efficacy. This review systematically summarizes metabolic reprogramming characteristics in both cancer and cardiovascular diseases, analyzes how anticancer therapies induce cardiovascular toxicity through metabolic alterations, and explores emerging therapeutic strategies targeting metabolic dysregulation. By integrating current research advancements, this review aims to enhance the understanding of CTR-CVT and provide groundwork for the development of safer and more effective cancer approaches.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Ying Gao
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China.
| | - Bo Lan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Jiani Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
34
|
Wang BD, Lucero A, Ha S, Yarmohammadi R. PI3Kδ as a Novel Therapeutic Target for Aggressive Prostate Cancer. Cancers (Basel) 2025; 17:1610. [PMID: 40427108 DOI: 10.3390/cancers17101610] [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: 03/23/2025] [Revised: 04/30/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) signaling represents an important pathway regulating cell proliferation, survival, invasion, migration, and metabolism. Notably, PI3K/AKT/mTOR signaling is frequently dysregulated in the majority of malignancies. Among the class IA PI3Ks (PI3Kα/β/δ), emerging evidence has implicated that PI3Kδ is not only overexpressed in leukocytes but also in solid tumors, including prostate cancer. The critical role of PI3Kδ in tumorigenesis and in the creation of a suppressive tumor microenvironment, along with the recent finding of PI3Kδ splice isoforms in promoting tumor aggressiveness and resistance, further demonstrates the potential of developing novel PI3Kδ-targeted cancer therapies. In this review, we comprehensively describe the functional mechanisms underlying the PI3Kδ-driven tumor progression and immune regulation in prostate cancer diseases. Furthermore, the recent preclinical and clinical studies on the development of PI3Kδ-/PI3K-targeted inhibitors as single agents and in combination therapies (with chemotherapy, radiation, hormone therapy, or immunotherapy) are summarized. Finally, we discuss the potential novel therapies for improving the treatment efficacies, as well as the current limitations and challenges of PI3Kδ-based therapies for prostate cancer.
Collapse
Affiliation(s)
- Bi-Dar Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA
- Hormone Related Cancers Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| | - Alyssa Lucero
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA
| | - Siyoung Ha
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA
| | - Reyhaneh Yarmohammadi
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA
| |
Collapse
|
35
|
Fu R, Li Z, Liu J, Xu B, Wen X, Zhang L. Potential mechanism of inhibitory effect of "medicine food homology" curcumin and its analogue EF24 on oral squamous cell carcinoma. Clin Transl Oncol 2025:10.1007/s12094-025-03871-8. [PMID: 40314923 DOI: 10.1007/s12094-025-03871-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/04/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumors of head and neck with high incidence and poor prognosis. Curcumin, as a drug-food congener, has a broad spectrum of anticancer effects, and based on this property, we further focused on EF24, a small molecule compound using curcumin as a backbone, to study the effects of both in OSCC. METHODS Cell experiments were performed to test the inhibitory effect of curcumin and EF24 on OSCC cells. The potential mechanism was further analyzed by transcriptome sequencing, and the DEGs after drug treatment were determined. PPI networks were created using Cytoscape software. RESULTS Both curcumin and EF24 inhibit the viability, migration, and invasion, and induce apoptosis of OSCC cells and the IC50 of EF24 was much lower than that of curcumin. Analysis of DEGs identified 893 DEGs following curcumin treatment, of which 794 were up-regulated and 99 were down-regulated; 797 DEGs following EF24 treatment were identified, of which 665 were up-regulated and 132 were down-regulated. Curcumin and EF24 were found to down-regulate lipid metabolism by key enzymes that regulate fatty acid and cholesterol synthesis. Furthermore, the number of T cell CD4 + memory is up-regulated and the immune response is enhanced. CONCLUSIONS It is suggested that curcumin and EF24 inhibit the metabolic reprogramming of tumor cells and at the same time regulate TME, and improve the immunotherapy of tumors, which opens the way for the future treatment of OSCC with this approach alone or in conjunction with immune-checkpoint blocking.
Collapse
Affiliation(s)
- Rao Fu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Huangpu District, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Huangpu District, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Ji'an Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Huangpu District, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Bo Xu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Huangpu District, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Xutao Wen
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Huangpu District, Shanghai, People's Republic of China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- National Center for Stomatology, Shanghai, People's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Ling Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Huangpu District, Shanghai, People's Republic of China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
- National Center for Stomatology, Shanghai, People's Republic of China.
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China.
- Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China.
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai, People's Republic of China.
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China.
- Kashagar District Second People's Hospital, Xinjiang, People's Republic of China.
| |
Collapse
|
36
|
Yu B, Cao Y, Lin P, Zhang L, Chen M. Enhancement of Ndrg2 promotes hypertrophic scar fibrosis by regulating PI3K/AKT signaling pathway. Cell Signal 2025; 129:111659. [PMID: 39956247 DOI: 10.1016/j.cellsig.2025.111659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/17/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Hypertrophic scar (HTS) is a prevalent chronic inflammatory skin disorder characterized by abnormal proliferation and extracellular matrix deposition. N-Myc downstream regulated gene 2 (Ndrg2) is a cell stress response gene related to cell proliferation, differentiation and various fibrotic diseases. However, the role of Ndrg2 in HTS is unknown and warrants further investigation. In this study, we confirmed that the expression of Ndrg2 was increased in HTS of human and a bleomycin-induced fibrosis mouse model. We then used Ndrg2 knockout mice and found Ndrg2 deletion could significantly reduce the synthesis of collagen and alleviate skin fibrosis. In addition, the proliferation and migration of Ndrg2-interfered HTS-derived fibroblasts decreased and those of Ndrg2-overexpressed normal skin-derived fibroblasts increased. Further, by western blot analysis, we verified that the expression of phosphorylated-PI3K, PI3K, phosphorylated-AKT and AKT were all increased after Ndrg2 overexpressed in normal skin-derived fibroblasts. Moreover, PI3K inhibitor (LY294002) administration significantly rescued the effect of Ndrg2 overexpression on skin fibrosis. In summary, our results demonstrated that Ndrg2 could promote HTS fibrosis by mediating PI3K/AKT signaling pathway. Our data suggest that Ndrg2 may be a promising therapeutic target for HTS.
Collapse
Affiliation(s)
- Boya Yu
- Department of Plastic and Reconstructive Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; Chinese PLA Medical School, Beijing 100853, China.
| | - Yalei Cao
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Pianpian Lin
- Department of Plastic and Reconstructive Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; Chinese PLA Medical School, Beijing 100853, China
| | - Lixia Zhang
- Department of Plastic and Reconstructive Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; Chinese PLA Medical School, Beijing 100853, China.
| | - Minliang Chen
- Department of Plastic and Reconstructive Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; Chinese PLA Medical School, Beijing 100853, China.
| |
Collapse
|
37
|
Gould S, Herron A, Davis J, Phillips M, Chakrabarti M, Evans CE. Control of inflammatory lung injury and repair by metabolic signaling in endothelial cells. Curr Opin Hematol 2025; 32:157-167. [PMID: 39450949 PMCID: PMC11949724 DOI: 10.1097/moh.0000000000000848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
PURPOSE OF REVIEW Sepsis-induced inflammatory lung injury includes acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). There are currently no effective treatments for ALI/ARDS, but clinical outcomes could be improved by inhibiting lung injury and/or promoting post-sepsis vascular repair. In this review, we describe studies of endothelial cell metabolic pathways in sepsis-induced ALI/ARDS and vascular repair and identify areas of research that deserve attention in future studies. We also describe studies of metabolic interventions that aim to inhibit ALI/ARDS and/or promote post-sepsis vascular repair, including those that target endothelial cell metabolites, endothelial cell metabolic signaling pathways, and endothelial cell metabolism. RECENT FINDINGS Endothelial cells are integral to both the injury and repair phases of ALI/ARDS. During the injury phase of ALI/ARDS, lung endothelial cell survival decreases, and lung endothelial cell-to-endothelial cell (EC-EC) junctions are weakened. During the repair phase after sepsis-induced lung injury, lung endothelial cell proliferation and lung EC-EC junction reannealing occur. These crucial aspects of ALI/ARDS and post-sepsis vascular repair, that is, endothelial cell viability, growth, and junction integrity, are controlled by a myriad of metabolites and metabolic signaling pathways in endothelial cells. SUMMARY Metabolic signaling pathways in endothelial cells represent a novel class of putative targets for the prevention and treatment of sepsis-induced inflammatory lung injury. Therapies that target metabolic signaling in endothelial cells are currently being explored as potential treatments for sepsis-induced inflammatory lung injury.
Collapse
Affiliation(s)
- Seth Gould
- Biomedical Engineering Program, University of South Carolina
| | - Ansley Herron
- Department of Chemical Engineering, College of Engineering and Computing, University of South Carolina
| | - Jonathan Davis
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
| | - Mollie Phillips
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
| | - Mrinmay Chakrabarti
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
| | - Colin E. Evans
- Biomedical Engineering Program, University of South Carolina
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine
- Institute on Cardiovascular Disease Research, University of South Carolina, Columbia, South Carolina, USA
| |
Collapse
|
38
|
Wang J, Chen W, Li Q, Yang R, Lin X, Han P, Huang X, Hu H, Luo M. AKT1 E17K-Interacting lncRNA SVIL-AS1 Promotes AKT1 Oncogenic Functions by Preferentially Blocking AKT1 E17K Dephosphorylation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500919. [PMID: 40135844 PMCID: PMC12097031 DOI: 10.1002/advs.202500919] [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] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/03/2025] [Indexed: 03/27/2025]
Abstract
AKT1E17K is a gain-of-function mutation that constitutively activates the PI3K-AKT pathway. However, how AKT1E17K is regulated in cancer pathogenesis remains elusive. Here, RNA immunoprecipitation sequencing (RIP-seq) is performed to interrogate the AKT1E17K-interacting lncRNAs and identify that SVIL-AS1 preferentially binds to AKT1E17K rather than AKT1WT proteins. It is found that SVIL-AS1 enhances AKT1 phosphorylation and downstream signaling. SVIL-AS1 knockdown dramatically inhibits the growth of AKT1E17K cells in vitro and in vivo. Notably, AKT1 and SVIL-AS1 interaction is AKT1 phosphorylation-dependent. SVIL-AS1 also interacts with PPP2R2A, a subunit of phosphatase PP2A holoenzyme, and blocks the binding of PPP2R2A to AKT1E17K to prevent AKT1 dephosphorylation. Moreover, AKT1E17K cells are not effectively inhibited by the allosteric AKT inhibitor, whereas silencing SVIL-AS1 sensitizes AKT1E17K cells to AKT1 allosteric inhibitor, as well as the PI3Kα inhibitor. In breast cancer tissues, SVIL-AS1 is highly expressed and associated with p-AKT1 level and poor prognosis of patients. Together, the findings discover a novel lncRNA regulator of mutant oncoprotein which preferentially prevents AKT1E17K dephosphorylation. Targeting SVIL-AS1 may help to improve the responses to inhibitors of the PI3K-AKT pathway, especially in AKT1E17K mutant tumors.
Collapse
Affiliation(s)
- Jingyi Wang
- Department of Otolaryngology‐Head and Neck SurgerySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
| | - Wenying Chen
- Department of GastroenterologySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
| | - Qianying Li
- Department of Otolaryngology‐Head and Neck SurgerySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
| | - Ruiyi Yang
- Department of OncologySun Yat‐Sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Xiaorong Lin
- Diagnosis and Treatment Center of Breast DiseasesShantou Central HospitalShantou515031China
| | - Ping Han
- Department of Otolaryngology‐Head and Neck SurgerySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
| | - Xiaoming Huang
- Department of Otolaryngology‐Head and Neck SurgerySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
| | - Hai Hu
- Breast Cancer CenterZhejiang Cancer HospitalHangzhou Institute of MedicineChinese Academy of SciencesHangzhou310022China
| | - Man‐Li Luo
- Medical Research CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
| |
Collapse
|
39
|
Kim D, Kesavan R, Ryu K, Dey T, Marckx A, Menezes C, Praharaj PP, Morley S, Ko B, Soflaee MH, Tom HJ, Brown H, Vu HS, Tso SC, Brautigam CA, Lemoff A, Mettlen M, Mishra P, Cai F, Allen DK, Hoxhaj G. Mitochondrial NADPH fuels mitochondrial fatty acid synthesis and lipoylation to power oxidative metabolism. Nat Cell Biol 2025; 27:790-800. [PMID: 40258949 DOI: 10.1038/s41556-025-01655-4] [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: 08/09/2024] [Accepted: 03/12/2025] [Indexed: 04/23/2025]
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) is a vital electron donor essential for macromolecular biosynthesis and protection against oxidative stress. Although NADPH is compartmentalized within the cytosol and mitochondria, the specific functions of mitochondrial NADPH remain largely unexplored. Here we demonstrate that NAD+ kinase 2 (NADK2), the principal enzyme responsible for mitochondrial NADPH production, is critical for maintaining protein lipoylation, a conserved lipid modification necessary for the optimal activity of multiple mitochondrial enzyme complexes, including the pyruvate dehydrogenase complex. The mitochondrial fatty acid synthesis (mtFAS) pathway utilizes NADPH for generating protein-bound acyl groups, including lipoic acid. By developing a mass-spectrometry-based method to assess mammalian mtFAS, we reveal that NADK2 is crucial for mtFAS activity. NADK2 deficiency impairs mtFAS-associated processes, leading to reduced cellular respiration and mitochondrial translation. Our findings support a model in which mitochondrial NADPH fuels the mtFAS pathway, thereby sustaining protein lipoylation and mitochondrial oxidative metabolism.
Collapse
Affiliation(s)
- Dohun Kim
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rushendhiran Kesavan
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kevin Ryu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Trishna Dey
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Austin Marckx
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cameron Menezes
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prakash P Praharaj
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stewart Morley
- Donald Danforth Plant Science Center, St. Louis, MO, USA
| | - Bookyung Ko
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mona H Soflaee
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Harrison J Tom
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Harrison Brown
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hieu S Vu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shih-Chia Tso
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chad A Brautigam
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marcel Mettlen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prashant Mishra
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Feng Cai
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Doug K Allen
- Donald Danforth Plant Science Center, St. Louis, MO, USA
- United States Department of Agriculture, Agriculture Research Service, St. Louis, MO, USA
| | - Gerta Hoxhaj
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
40
|
Qi Z, Luo J, Liu W, Xu Y, Ma Y, Hu S, Shen X, Du X, Xiang W. Alisertib impairs the stemness of hepatocellular carcinoma by inhibiting purine synthesis. J Biol Chem 2025; 301:108558. [PMID: 40311679 DOI: 10.1016/j.jbc.2025.108558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 05/03/2025] Open
Abstract
Hepatocellular carcinoma tumor-repopulating cells (HCC-TRCs) drive disease progression, yet their purine metabolism mechanisms remain poorly understood. This study revealed that the stemness index, strongly linked to poor HCC prognosis, exhibited a robust positive correlation with purine metabolism through single-sample gene set enrichment analysis. Integrated drug screening across CTRP, GDSC, and PRISM databases identified alisertib, an aurora kinase A (AURKA) inhibitor, as a potent agent targeting stemness. Using fibrin gel-based 3D-cultured HCC-TRCs, mechanistic studies demonstrated that alisertib suppresses xanthine and hypoxanthine production by inhibiting the AURKA-AKT signaling axis. This disruption markedly impaired tumor spheroid formation, migration, and invasion in vitro, while significantly suppressed tumor growth in vivo, which could be rescued by the AKT agonist SC79. Our findings revealed a novel therapeutic strategy targeting purine metabolism through AURKA-AKT axis inhibition, effectively eliminating HCC-TRCs.
Collapse
Affiliation(s)
- Zhuoran Qi
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Luo
- Huashan Hospital, Fudan University, Shanghai, China
| | - Wenfeng Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ye Xu
- Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifan Ma
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sunkuan Hu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Xiaojing Du
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Wei Xiang
- Department of Interventional Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
41
|
Wang X, Qu Y, Sun Y, Yang T, Wang W, Dou X, Jia Y. ATP6V0B promotes the tumorigenesis of bladder cancer by activating PAQR4/PI3K/AKT signaling. BMC Cancer 2025; 25:789. [PMID: 40295930 PMCID: PMC12036214 DOI: 10.1186/s12885-025-14183-z] [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: 12/29/2024] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND ATPase H+ transporting V0 subunit b (ATP6V0B) is an essential component of the vacuolar ATP multi-protein complex (V-ATPase) associated with energy metabolism. However, information on its role and mechanism of action in bladder cancer (BCa) and other tumors is not clear. METHODS In this study, we evaluated the expression of ATP6V0B in BCa and its correlation with patient survival outcomes by performing public database analysis, as well as, RT-qPCR and Western blotting assays. We also investigated the effect of altering the level of expression of ATP6V0B on the malignant behavior of BCa cells at the cellular level by conducting the CCK-8 assay and Transwell assay. In vivo experiments involved subcutaneous injection of stable ATP6V0B-knockdown BCa cells into nude mice to assess the influence of ATP6V0B on tumorigenesis. Additionally, bioinformatics analysis was combined with other methods to predict that ATP6V0B may modulate signaling pathways. RESULTS The findings showed that the expression of ATP6V0B increased in BCa tissues, and patients exhibiting high levels of this protein had a poorer prognosis. Additionally, our results showed that ATP6V0B functions as an oncogene and stimulates the proliferation, invasion, and migration of BCa cells in vitro. In vivo animal studies showed that downregulating ATP6V0B hindered the growth of BCa. Regarding the mechanism of action of ATVP60VB, we found that ATVP60VB can activate the PI3K/AKT signaling pathway through Progestin and AdipoQ Receptor Family Member 4 (PAQR4) -mediated upregulation. CONCLUSION To summarize, the results of this study indicated that an increase in the level of expression of ATP6V0B in BCa tissues and cells is associated with unfavorable patient prognosis due to its tumor-promoting effects via upregulation of the PAQR4/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Xinsheng Wang
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300211, China
| | - Yanqing Qu
- Surgical Clinic, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, 266071, China
| | - Yanbo Sun
- Department of Urology, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, Shandong, 266000, China
| | - Tong Yang
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300211, China
| | - Wei Wang
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300211, China
| | - Xinmeng Dou
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300211, China
| | - Yong Jia
- Department of Urology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), No. 1 Jiaozhou Road, Qingdao, Shandong, 266071, China.
| |
Collapse
|
42
|
Yin S, Brobbey C, Ball LE, Fu T, Sprague DJ, Gan W. BRD9 functions as a methylarginine reader to regulate AKT-EZH2 signaling. SCIENCE ADVANCES 2025; 11:eads6385. [PMID: 40279411 PMCID: PMC12024519 DOI: 10.1126/sciadv.ads6385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 03/20/2025] [Indexed: 04/27/2025]
Abstract
Recognition of methylarginine marks by effector proteins ("readers") is a critical link between arginine methylation and various cellular processes. Recently, we identified methylation of AKT1 at arginine-391 (R391), but the reader for this methylation has yet to be characterized. Here, we show that bromodomain-containing protein 9 (BRD9), a reader of acetylated lysine, unexpectedly recognizes methylated R391 of AKT1 through an aromatic cage in its bromodomain. Disrupting the methylarginine reader function of BRD9 suppresses AKT activation and tumorigenesis. RNA sequencing data show that BRD9 and AKT coregulate a hallmark transcriptional program in part through enhancer of zeste homolog 2 (EZH2)-mediated methylation of histone-3 lysine-27. We also find that inhibitors of BRD9 and EZH2 display synergistic effects on suppression of cell proliferation and tumor growth. Collectively, our study reveals a previously unknown function of BRD9 and a potential therapeutic strategy for cancer treatment by combining BRD9 and EZH2 inhibitors.
Collapse
Affiliation(s)
- Shasha Yin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Charles Brobbey
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Lauren E. Ball
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Tianmin Fu
- Department of Biological Chemistry and Pharmacology, Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel J. Sprague
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Wenjian Gan
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
43
|
Sun J, Qi X, Yang C, Wang S, Jiang J, Wang L, Song J, Yu B, Sun M. Network Pharmacology, Molecular Docking, and in vitro Experiments Reveal the Role and Mechanism of Tanshinone IIA in Colorectal Cancer Treatment Through the PI3K/AKT Pathway. Drug Des Devel Ther 2025; 19:2959-2977. [PMID: 40255473 PMCID: PMC12009581 DOI: 10.2147/dddt.s492033] [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: 12/11/2024] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
Purpose To examine the roles and mechanisms of tanshinone IIA (Tan-IIA) in colorectal cancer (CRC) using network pharmacology, molecular docking, and in vitro experiments. Methods In network pharmacology studies, Tan-IIA targets for treating CRC were identified using public databases. Employing the protein-protein interaction (PPI) network, gene ontology (GO) enrichment, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, the core genes and mechanisms of action of Tan-IIA were obtained. Core targets were validated using Gene Expression Profiling Interactive Analysis, the Human Protein Atlas, DriverDBv3, cBioPortal, and the Tumor Immune Estimation Resource database. Molecular docking validates the binding affinity of Tan-IIA to some key targets. Network pharmacology and molecular docking results were validated via in vitro experiments. Results Intersecting Tan-IIA and CRC targets led to the identification of 25 potential targets. PPI analysis identified 10 core targets of Tan-IIA for CRC treatment. Database validation revealed that these core targets were expressed at varying levels in both normal and cancer tissues. Their expression could influence patient prognosis and immune cell infiltration levels. GO analysis revealed 170 biological processes, 42 cellular components, and 83 molecular functions. KEGG analysis indicated that Tan-IIA affected CRC through multiple pathways, including the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT), cAMP, and TNF signaling pathways, with the PI3K/AKT pathway being the most enriched. Molecular docking results indicated that Tan-IIA effectively binds to PI3K, AKT, and other partial core targets. In vitro experiments revealed that Tan-IIA suppressed the multiplication and migration of HCT116 and SW480 cells, induced apoptosis, and reduced the PI3K/AKT pathway indicator protein expression, which was reversed by the PI3K/AKT pathway agonist insulin-like growth factor-1. Conclusion Network pharmacology, molecular docking, and in vitro validation confirmed that Tan-IIA contributes to CRC treatment through the PI3K/AKT pathway, providing theoretical and experimental foundations for its potential clinical application.
Collapse
Affiliation(s)
- Jinpeng Sun
- College of Integrative Chinese and Western Medicine, Jining Medical University, Jining, Shandong, 272067, People’s Republic of China
| | - Xinmeng Qi
- College of Integrative Chinese and Western Medicine, Jining Medical University, Jining, Shandong, 272067, People’s Republic of China
| | - Cuiyuan Yang
- College of Integrative Chinese and Western Medicine, Jining Medical University, Jining, Shandong, 272067, People’s Republic of China
| | - Shanpeng Wang
- College of Integrative Chinese and Western Medicine, Jining Medical University, Jining, Shandong, 272067, People’s Republic of China
| | - Jingwen Jiang
- College of Integrative Chinese and Western Medicine, Jining Medical University, Jining, Shandong, 272067, People’s Republic of China
| | - Lijie Wang
- College of Integrative Chinese and Western Medicine, Jining Medical University, Jining, Shandong, 272067, People’s Republic of China
| | - Jiacheng Song
- College of Integrative Chinese and Western Medicine, Jining Medical University, Jining, Shandong, 272067, People’s Republic of China
| | - Bin Yu
- College of Integrative Chinese and Western Medicine, Jining Medical University, Jining, Shandong, 272067, People’s Republic of China
| | - Min Sun
- College of Integrative Chinese and Western Medicine, Jining Medical University, Jining, Shandong, 272067, People’s Republic of China
| |
Collapse
|
44
|
Zhao Y, Xu H, Liu Q, Yuan Y, Li R, Li D, Zhang Y, Ran J, Yan X, Su J. The interaction between IL-33 and TRIM28 in the regulation of macrophage polarization in an ST2-independent manner. Int Immunopharmacol 2025; 152:114318. [PMID: 40054323 DOI: 10.1016/j.intimp.2025.114318] [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: 11/12/2024] [Revised: 01/24/2025] [Accepted: 02/13/2025] [Indexed: 03/24/2025]
Abstract
The tumor microenvironment provides optimal condition for the growth of ovarian cancer. Macrophages display a highly functional plasticity to respond various signals. Switching macrophages' phenotype is a potential therapeutic strategy for the treatment of cancer. We used RNA-sequencing(RNA-Seq) and Chromatin immunoprecipitation-sequencing(ChIP-Seq) analyses in bone-marrow-derived macrophages (BMDMs) from wild-type (WT) and its receptor interleukin-1 receptor like-1 (IL1RL1 or ST2) knockout(ST2-/-) mice revealed that the interaction between IL-33 and TRIM28, which plays an antioxidant role, regulates glycolysis in BMDMs by promoting the PI3K/Akt pathway in ST2-independent manner, thereby reducing M2 polarization of macrophages is a way to inhibit ovarian cancer growth.
Collapse
Affiliation(s)
- Yuanxin Zhao
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China.
| | - Huadan Xu
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China.
| | - Qingqing Liu
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China.
| | - Yuan Yuan
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China.
| | - Runyuan Li
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China.
| | - Dong Li
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China.
| | - Yong Zhang
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China.
| | - Jingyi Ran
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Xiaoyu Yan
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China.
| | - Jing Su
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China.
| |
Collapse
|
45
|
Shin AE, Sugiura K, Kariuki SW, Cohen DA, Flashner SP, Klein-Szanto AJ, Nishiwaki N, De D, Vasan N, Gabre JT, Lengner CJ, Sims PA, Rustgi AK. LIN28B-mediated PI3K/AKT pathway activation promotes metastasis in colorectal cancer models. J Clin Invest 2025; 135:e186035. [PMID: 39808497 PMCID: PMC11996871 DOI: 10.1172/jci186035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer death because of metastatic spread. LIN28B is overexpressed in 30% of CRCs and promotes metastasis, yet its mechanisms remain unclear. In this study, we genetically modified CRC cell lines to overexpress LIN28B, resulting in enhanced PI3K/AKT pathway activation and liver metastasis in mice. We developed genetically modified mouse models with constitutively active Pik3ca that form intestinal tumors progressing to liver metastases with an intact immune system, addressing the limitations of previous Pik3ca-mutant models, including long tumor latency, mixed histology, and lack of distant metastases. The PI3Kα-specific inhibitor alpelisib reduced migration and invasion in vitro and metastasis in vivo. We present a comprehensive analysis of vertical inhibition of the PI3K/AKT pathway in CRC using the FDA-approved drugs alpelisib and capivasertib (an AKT inhibitor) in combination with LY2584702 (a ribosomal protein S6 kinase inhibitor) in CRC cell lines and mouse- and patient-derived organoids. Tissue microarrays from patients with CRC verified that LIN28B and PI3K/AKT pathway activation correlate with CRC progression. These findings highlight the critical role of the LIN28B-mediated PI3K/AKT pathway in CRC metastasis, the therapeutic potential of targeted inhibition, and the promise of patient-derived organoids in precision medicine in metastatic CRC.
Collapse
Affiliation(s)
- Alice E. Shin
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Kensuke Sugiura
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | | | - David A. Cohen
- Department of Surgery, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons; Columbia University Irving Medical Center, New York, New York, USA
| | | | | | | | - Dechokyab De
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Neil Vasan
- Division of Hematology and Oncology, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Joel T. Gabre
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Christopher J. Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter A. Sims
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Anil K. Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, and
| |
Collapse
|
46
|
Omezzolli G, Iannello A, Vallone FE, Brandimarte L, Micillo M, Bertola N, Lavarello C, Grinovero N, Ferrero G, Mellert K, Möller P, Bruno S, Furman RR, Allan JN, Petretto A, Deaglio S, Ravera S, Vaisitti T. Complementary approaches define the metabolic features that accompany Richter syndrome transformation. Cell Mol Life Sci 2025; 82:152. [PMID: 40204982 PMCID: PMC11982009 DOI: 10.1007/s00018-025-05670-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/25/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
Richter syndrome (RS) is the transformation of chronic lymphocytic leukemia (CLL) into a high-grade lymphoma with previously unknown metabolic features. Transcriptomic data from primary CLL and RS samples, as well as RS-patient-derived xenografts, highlighted cellular metabolism as one of the most significant differentially expressed processes. Activity assays of key enzymes confirmed the intense metabolic rewiring of RS cells, which is characterized by an elevated rate of Krebs cycle, oxidative phosphorylation, and glutamine metabolism. These pathways were sustained by increased uptake of glucose and glutamine, two critical substrates for these cells. Moreover, RS cells showed activation of anabolic processes that resulted in the synthesis of nucleotides and lipids necessary to support their high proliferation. Exposure to drugs targeting PI3K and NF-kB, two master regulators of cellular metabolism, resulted in the shutdown of ATP production and glycolysis. Overall, these data suggest that metabolic rewiring characterizes the transformation of CLL into RS, presenting new translational opportunities.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Animals
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cell Transformation, Neoplastic/genetics
- Glycolysis
- Citric Acid Cycle
- Mice
- Oxidative Phosphorylation
- Glucose/metabolism
- Glutamine/metabolism
- NF-kappa B/metabolism
- NF-kappa B/antagonists & inhibitors
- Phosphatidylinositol 3-Kinases/metabolism
Collapse
Affiliation(s)
- Giulia Omezzolli
- Department of Medical Sciences, University of Torino, Via Nizza 52, 10126, Turin, Italy
| | - Andrea Iannello
- Department of Medical Sciences, University of Torino, Via Nizza 52, 10126, Turin, Italy
| | - Francesco E Vallone
- Department of Medical Sciences, University of Torino, Via Nizza 52, 10126, Turin, Italy
| | - Lorenzo Brandimarte
- Department of Medical Sciences, University of Torino, Via Nizza 52, 10126, Turin, Italy
| | - Matilde Micillo
- Department of Medical Sciences, University of Torino, Via Nizza 52, 10126, Turin, Italy
| | - Nadia Bertola
- U.O. Molecular Pathology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Lavarello
- Core Facilities-Clinical Proteomics and Metabolomics, IRCCS Giannina Gaslini, Genoa, Italy
| | - Nicole Grinovero
- Core Facilities-Clinical Proteomics and Metabolomics, IRCCS Giannina Gaslini, Genoa, Italy
| | - Giulio Ferrero
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Kevin Mellert
- Institute of Pathology, University Hospital Ulm, Ulm, Germany
| | - Peter Möller
- Institute of Pathology, University Hospital Ulm, Ulm, Germany
| | - Silvia Bruno
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Richard R Furman
- Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY, USA
| | - John N Allan
- Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY, USA
| | - Andrea Petretto
- Core Facilities-Clinical Proteomics and Metabolomics, IRCCS Giannina Gaslini, Genoa, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Torino, Via Nizza 52, 10126, Turin, Italy
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Tiziana Vaisitti
- Department of Medical Sciences, University of Torino, Via Nizza 52, 10126, Turin, Italy.
| |
Collapse
|
47
|
Zhang Y, Jin H, Jia W, Liu Y, Wang Y, Xue S, Liu Y, Hao H. Ermiao San attenuating rheumatoid arthritis via PI3K/AKT/mTOR signaling activate HIF-1α induced glycolysis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119615. [PMID: 40081512 DOI: 10.1016/j.jep.2025.119615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/14/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a classic formula, Ermiao San (EMS) characterized by its less medicinal flavor and strong potency had been proven to be effective and safe in the treatment of rheumatoid arthritis (RA) during clinical experience and our previous research. AIM OF THE STUDY The therapeutic characteristics of multi-component and multi-target of traditional Chinese medicine prompted us to further investigated the effective compounds of EMS, and evaluated its potential mechanisms in treating RA. MATERIALS AND METHODS Ultra-high-performance liquid chromatography high-resolution mass spectrometry (UPLC-HRMS) was used to analyze the primary absorption components of EMS in rat serum, with secondary mass spectrometry used to assist in identifying the structures of the compounds. Open field experiments, H&E staining, safranin-O-turquoise staining, ELISA, and other methods were applied to verify the alleviating effects of EMS on exercise capacity, inflammation, and cartilage damage in CIA rats. The RA-FLS model was established using TNF-α, and observed the effects of EMS on cell migration and invasion were observed through wound healing and transwell assays. In addition, immunohistochemistry and western blotting were employed to investigate the PI3K/AKT/mTOR/HIF-1α pathway both in vivo and in vitro. RESULTS Seventeen compounds were identified in rat serum, which were considered as active ingredients involved in the improvement of RA by EMS. Furthermore, EMS demonstrated the outstanding anti-RA ability, as evidenced by the improvement in foot swelling and arthritis scores, alleviation of pathological changes in joint tissue, inhibition of inflammatory factors, and restoration of exercise ability. In vivo data showed that EMS reduced joint injury through the PI3K/AKT/mTOR/HIF-1α signaling pathway. In vitro studies indicated that TNF-α induced the expression of Glut1 and HK2 proteins, accelerated the glycolysis rate, and promoted migration and invasion of RA-FLS cells, leading to adverse outcomes. However, EMS regulated the expression of glycolysis-related molecules, HK2 and Glut1 through the PI3K/AKT/mTOR/HIF-1α pathway, thereby inhibiting inflammation, migration, and invasion of RA-FLS cells. CONCLUSION The beneficial effects of EMS in CIA rats can be attributed to the inhibition of glycolysis in synovial fibroblasts via the PI3K/AKT/mTOR/HIF-1α pathway. This finding further enriches our understanding of the mechanisms by which EMS contributes to the treatment of RA.
Collapse
Affiliation(s)
- Yumeng Zhang
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Basic Laboratory of Integrated Traditional Chinese and Western Medicine, Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Engineering Research Center of Cross Innovation for Chinese Traditional Medicine of Shanxi Province, Jinzhong, 030619, China
| | - Haizhu Jin
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Wenyue Jia
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yuqi Liu
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yuru Wang
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Shuyan Xue
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Yang Liu
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Basic Laboratory of Integrated Traditional Chinese and Western Medicine, Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Engineering Research Center of Cross Innovation for Chinese Traditional Medicine of Shanxi Province, Jinzhong, 030619, China.
| | - Huiqin Hao
- Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Basic Laboratory of Integrated Traditional Chinese and Western Medicine, Shanxi University of Chinese Medicine, Jinzhong, 030619, China; Engineering Research Center of Cross Innovation for Chinese Traditional Medicine of Shanxi Province, Jinzhong, 030619, China.
| |
Collapse
|
48
|
Mehmood A, Hakami MA, Ogaly HA, Subramaniyan V, Khalid A, Wadood A. Evolution of computational techniques against various KRAS mutants in search for therapeutic drugs: a review article. Cancer Chemother Pharmacol 2025; 95:52. [PMID: 40195161 DOI: 10.1007/s00280-025-04767-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/23/2025] [Indexed: 04/09/2025]
Abstract
KRAS was (Kirsten rat sarcoma viral oncogene homolog) revealed as an important target in current therapeutic cancer research because alteration of RAS (rat sarcoma viral oncogene homolog) protein has a critical role in malignant modification, tumor angiogenesis, and metastasis. For cancer treatment, designing competitive inhibitors for this attractive target was difficult. Nevertheless, computational investigations of the protein's dynamic behavior displayed the existence of temporary pockets that could be used to design allosteric inhibitors. The last decade witnessed intensive efforts to discover KRAS inhibitors. In 2021, the first KRAS G12C covalent inhibitor, AMG 510, received FDA (Food and drug administration) approval as an anticancer medication that paved the path for future treatment strategies against this target. Computer-aided drug designing discovery has long been used in drug development research targeting different KRAS mutants. In this review, the major breakthroughs in computational methods adapted to discover novel compounds for different mutations have been discussed. Undoubtedly, virtual screening and molecular dynamic (MD) simulation and molecular docking are the most considered approach, producing hits that can be employed in subsequent refinements. After comprehensive analysis, Afatinib and Quercetin were computationally identified as hits in different publications. Several authors conducted covalent docking studies with acryl amide warheads groups containing inhibitors. Future studies are needed to demonstrate their true potential. In-depth studies focusing on various allosteric pockets demonstrate that the switch I/II pocket is a suitable site for drug designing. In addition, machine learning and deep learning based approaches provide new insights for developing anti-KRAS drugs. We believe that this review provides extensive information to researchers globally and encourages further development in this particular area of research.
Collapse
Affiliation(s)
- Ayesha Mehmood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al- Quwayiyah, Riyadh, Saudi Arabia
| | - Hanan A Ogaly
- Chemistry Department, College of Science, King Khalid University, Abha, 61421, Saudi Arabia
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, School of Medical and Life Sciences, Sunway University No. 5, Jalan Universiti, Bandar Sunway, Selangor Darul Ehsan, 47500, Malaysia
| | - Asaad Khalid
- Health Research Center, Jazan University, 114, Jazan, 45142, Saudi Arabia
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan.
| |
Collapse
|
49
|
Dastgheib SA, Bahrami R, Golshan-Tafti M, Danaei M, Azizi S, Shahbazi A, Yeganegi M, Shiri A, Masoudi A, Neamatzadeh H. Decoding bronchopulmonary dysplasia in premature infants through an epigenetic lens. Front Med (Lausanne) 2025; 12:1531169. [PMID: 40248086 PMCID: PMC12003331 DOI: 10.3389/fmed.2025.1531169] [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: 11/19/2024] [Accepted: 02/05/2025] [Indexed: 04/19/2025] Open
Abstract
This review provides a comprehensive overview of the evolving insights into the epigenetic mechanisms associated with bronchopulmonary dysplasia (BPD). It specifically highlights the roles of DNA methylation, histone modifications, and RNA regulation in the development of BPD in premature infants. BPD results from complex interactions among genetic factors, environmental exposures, and neonatal stressors. Key findings suggest that intrauterine hypoxia, hyperoxia, and nutrition can lead to epigenetic alterations, affecting gene expression and methylation, which may serve as biomarkers for early BPD detection. RUNX3 is identified as a critical transcription factor influencing lung development and inflammation, while changes in DNA methylation and histone dynamics in cord blood are linked to immune dysregulation associated with BPD. The role of m6A RNA methylation regulators from the IGF2BP family affects mRNA stability and gene expression relevant to BPD. Additionally, specific histones and microRNAs, particularly from the miR-17∼92 cluster, are implicated in pulmonary development and vascular regulation. Long non-coding RNAs (lncRNAs), such as MALAT1, also play a role in gene regulation via competitive endogenous RNA networks, indicating their potential as biomarkers and therapeutic targets. The interplay of these epigenetic mechanisms underscores the need for further research to develop targeted interventions aimed at reducing BPD severity and enhancing health outcomes for at-risk neonates.
Collapse
Affiliation(s)
- Seyed Alireza Dastgheib
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Bahrami
- Neonatal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mahsa Danaei
- Department of Obstetrics and Gynecology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Azizi
- Shahid Akbarabadi Clinical Research Development Unit, Iran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Yeganegi
- Department of Obstetrics and Gynecology, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Amirmasoud Shiri
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Masoudi
- School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Neamatzadeh
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
50
|
Llorente A, Arora GK, Murad R, Emerling BM. Phosphoinositide kinases in cancer: from molecular mechanisms to therapeutic opportunities. Nat Rev Cancer 2025:10.1038/s41568-025-00810-1. [PMID: 40181165 DOI: 10.1038/s41568-025-00810-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/28/2025] [Indexed: 04/05/2025]
Abstract
Phosphoinositide kinases, extending beyond the well-known phosphoinositide 3-kinase (PI3K), are key players in the dynamic and site-specific phosphorylation of lipid phosphoinositides. Unlike PI3Ks, phosphatidylinositol 4-kinases (PI4Ks) and phosphatidylinositol phosphate kinases (PIPKs) do not usually exhibit mutational alterations, but mostly show altered expression in tumours, orchestrating a broad spectrum of signalling, metabolic and immune processes, all of which are crucial in the pathogenesis of cancer. Dysregulation of PI4Ks and PIPKs has been associated with various malignancies, which has sparked considerable interest towards their therapeutic targeting. In this Review we summarize the current understanding of the lesser-studied phosphoinositide kinase families, PI4K and PIPK, focusing on their functions and relevance in cancer. In addition, we provide an overview of ongoing efforts driving the preclinical and clinical development of phosphoinositide kinase-targeting molecules.
Collapse
Affiliation(s)
- Alicia Llorente
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Gurpreet K Arora
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Rabi Murad
- Bioformatics Core, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Brooke M Emerling
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|