1
|
Albassam H, Almutairi O, Alnasser M, Altowairqi F, Almutairi F, Alobid S. Discovery of a selective PI3Kα inhibitor via structure-based virtual screening for targeted colorectal cancer therapy. J Enzyme Inhib Med Chem 2025; 40:2468852. [PMID: 39992303 PMCID: PMC11852364 DOI: 10.1080/14756366.2025.2468852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality globally, driving an urgent need for effective therapies. A promising avenue of research focuses on the PI3K/AKT/mTOR signalling pathway, which is frequently disrupted by mutations in the PI3Kα subunit. Our cutting-edge study employed a structure-based virtual screening of ∼3000 compounds, leading to the discovery of F0608-0019, a highly potent and selective PI3Kα inhibitor. F0608-0019 demonstrated remarkable efficacy in suppressing HCT116 colorectal cancer cell proliferation, with an IC50 of 12.14 µM, while maintaining high selectivity by minimising activity against other PI3K isoforms. Advanced molecular dynamics simulations highlighted the stability of F0608-0019's binding interactions with key amino acids, such as TRP:780, ILE:932, and VAL:850, which are critical for its targeted action. These exciting findings reveal F0608-0019 as a leading candidate for innovative CRC therapies that selectively target PI3Kα dysregulation, offering promising new possibilities for effective CRC treatment.
Collapse
Affiliation(s)
- Hussam Albassam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Omar Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Majed Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faisal Altowairqi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faris Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saad Alobid
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Saadh MJ, Allela OQB, Kareem RA, Baldaniya L, Ballal S, Vashishth R, Parmar M, Sameer HN, Hamad AK, Athab ZH, Adil M. Prognostic gene expression profile of colorectal cancer. Gene 2025; 955:149433. [PMID: 40122415 DOI: 10.1016/j.gene.2025.149433] [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: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer is a major global health burden, with significant heterogeneity in clinical outcomes among patients. Identifying robust prognostic gene expression signatures can help stratify patients, guide treatment decisions, and improve clinical management. This review provides an overview of current prognostic gene expression profiles in colorectal cancer research. We have synthesized evidence from numerous published studies investigating the association between tumor gene expression patterns and patient survival outcomes. The reviewed literature reveals several promising gene signatures that have demonstrated the ability to predict disease-free survival and overall survival in CRC patients, independent of standard clinicopathological risk factors. These genes are crucial in fundamental biological processes, including cell cycle control, epithelial-mesenchymal transition, and immune regulation. The implementation of prognostic gene expression tests in clinical practice holds great potential for enabling more personalized management strategies for colorectal cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Manisha Parmar
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India.
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq.
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq.
| | | |
Collapse
|
3
|
Richter EA, Bilan PJ, Klip A. A comprehensive view of muscle glucose uptake: regulation by insulin, contractile activity, and exercise. Physiol Rev 2025; 105:1867-1945. [PMID: 40173020 DOI: 10.1152/physrev.00033.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/07/2024] [Accepted: 03/08/2025] [Indexed: 04/04/2025] Open
Abstract
Skeletal muscle is the main site of glucose deposition in the body during meals and the major glucose utilizer during physical activity. Although in both instances the supply of glucose from the circulation to the muscle is of paramount importance, in most conditions the rate-limiting step in glucose uptake, storage, and utilization is the transport of glucose across the muscle cell membrane. This step is dependent upon the translocation of the insulin- and contraction-responsive glucose transporter GLUT4 from intracellular storage sites to the sarcolemma and T tubules. Here, we first analyze how glucose can traverse the capillary wall into the muscle interstitial space. We then review the molecular processes that regulate GLUT4 translocation in response to insulin and muscle contractions and the methodologies utilized to unravel them. We further discuss how physical activity and inactivity, respectively, lead to increased and decreased insulin action in muscle and touch upon sex differences in glucose metabolism. Although many key processes regulating glucose uptake in muscle are known, the advent of newer and bioinformatics tools has revealed further molecular signaling processes reaching a staggering level of complexity. Much of this molecular mapping has emerged from cellular and animal studies and more recently from application of a variety of -omics in human tissues. In the future, it will be imperative to validate the translatability of results drawn from experimental systems to human physiology.
Collapse
Affiliation(s)
- Erik A Richter
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Philip J Bilan
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Lu Z, Wang T, Wang L, Ming J. Research progress on estrogen receptor-positive/progesterone receptor-negative breast cancer. Transl Oncol 2025; 56:102387. [PMID: 40222338 PMCID: PMC12018574 DOI: 10.1016/j.tranon.2025.102387] [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: 10/07/2024] [Revised: 03/19/2025] [Accepted: 04/05/2025] [Indexed: 04/15/2025] Open
Abstract
Breast cancer, which arises from the epithelial tissue of the breast, is one of the most common cancers affecting women worldwide. Its incidence and mortality rates have been increasing in both developed and developing countries. As a hormone-dependent cancer, breast cancer is classified into several molecular subtypes based on the expression of key markers: Estrogen Receptor (ER), Progesterone Receptor (PR), Human Epidermal Growth Factor Receptor 2 (HER-2), and Ki67. PR loss is associated with endocrine resistance and a poorer prognosis in breast cancer. Despite this, the underlying mechanisms of ER-positive/PR-negative (ER+PR-) breast cancer remain poorly understood. This study aims to review recent advancements in research on ER+PR- breast cancer, analyze its clinical characteristics and molecular mechanisms, and provide recommendations for more targeted therapeutic approaches.
Collapse
Affiliation(s)
- Zhengjia Lu
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tingrui Wang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lu Wang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jia Ming
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
5
|
Zhang C, Zhang S, Wang G, Huang X, Xu S, Wang D, Guo C, Wang Y. Genomics and transcriptomics identify quantitative trait loci affecting growth-related traits in silver pomfret (Pampus argenteus). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101414. [PMID: 39813916 DOI: 10.1016/j.cbd.2025.101414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
Pampus argenteus, a species distributed throughout the Indo-West Pacific, plays a significant role in the yield of aquaculture species. However, cultured P. argenteus has always been characterised by unbalanced growth synchronisation among individuals, slow growth rate, and lack of excellent germplasm resources. Therefore, we conducted mass selection for fast-growing strain P. argenteus for several consecutive years. Various genetic improvement programs have modified its genome sequence through selective pressure, leaving nucleotide signals that can be detected at the genomic level. In the present study, we combined bulked segregant analysis and transcriptome sequencing to identify candidate single nucleotide polymorphisms (SNPs) and key genes for growth-related traits in P. argenteus. A total of 7,280,936 SNPs and 2,212,379 insertions/deletions were identified in the extreme phenotypes of the fast-growing and slow-growing groups. Based on the examination of SNP frequency differences and sliding-window analysis, 42 SNPs were identified as candidate markers. Moreover, 14 of the 42 SNPs linked to growth-related traits were confirmed to be credible SNPs, and eight growth-related genes were screened, namely myb-binding protein 1 A, insulin A/B chains, α-1B adrenoceptor, engulfment and cell motility protein 3, myosin light chain kinase family member 4, insulin receptor located, unconventional myosin-9b, and matrilin-1. An optimal three-factor model (SNP4&SNP12&SNP14) was constructed using the generalized multifactor dimensionality reduction method, and its accuracy was verified as 67.72 %. These results may benefit genetic studies and accelerate genetic improvement of fast-growing strains of P. argenteus.
Collapse
Affiliation(s)
- Cheng Zhang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China; College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Shun Zhang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Guanlin Wang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Xiang Huang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Shanliang Xu
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Danli Wang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Chunyang Guo
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Zhejiang, Ningbo 315211, China.
| | - Yajun Wang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Zhejiang, Ningbo 315211, China.
| |
Collapse
|
6
|
Liu L, Wuyun T, Sun X, Zhang Y, Cha G, Zhao L. Therapeutic efficacy of TMTP1-modified EVs in overcoming bone metastasis and immune resistance in PIK3CA mutant NSCLC. Cell Death Dis 2025; 16:367. [PMID: 40328748 PMCID: PMC12055990 DOI: 10.1038/s41419-025-07685-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025]
Abstract
Non-small cell lung cancer (NSCLC) with PIK3CA mutations demonstrates significant challenges in treatment due to enhanced bone metastasis and immune checkpoint resistance. This study investigates the efficacy of tumor-targeting peptide 1-modified cancer stem cell-derived extracellular vesicles (TMTP1-TSRP-EVs) in reshaping the tumor microenvironment and reversing immune checkpoint resistance in NSCLC. By integrating TMTP1-TSRP into EVs, we aim to specifically deliver therapeutic agents to NSCLC cells, focusing on inhibiting the PI3K/Akt/mTOR pathway, a crucial driver of oncogenic activity and immune evasion in PIK3CA-mutated cells. Our comprehensive in vitro and in vivo analyses show that TMTP1-TSRP-EVs significantly inhibit tumor growth, reduce PD-L1 expression, and enhance CD8+ T cell infiltration, effectively reversing the immune-suppressive microenvironment. Moreover, the in vivo models confirm that our approach not only suppresses bone metastases but also overcomes primary resistance to immune checkpoint inhibitors by modulating the expression of key immunological markers. These findings suggest that targeted delivery of TMTP1-TSRP-EVs could provide a novel therapeutic strategy for treating PIK3CA-mutant NSCLC, offering significant improvements over traditional therapies by directly targeting the molecular pathogenesis of tumor resistance and metastasis. Molecular Mechanisms Reshaping the TME to Halt PI3K-Mutant Bone Metastasis of NSCLC and Overcoming Primary ICI Resistance. (Created by BioRender).
Collapse
Affiliation(s)
- Liwen Liu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tanghesi Wuyun
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Sun
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Zhang
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Geqi Cha
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ling Zhao
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
7
|
Hedayati N, Safari MH, Milasi YE, Kahkesh S, Farahani N, Khoshnazar SM, Dorostgou Z, Alaei E, Alimohammadi M, Rahimzadeh P, Taheriazam A, Hashemi M. Modulation of the PI3K/Akt signaling pathway by resveratrol in cancer: molecular mechanisms and therapeutic opportunity. Discov Oncol 2025; 16:669. [PMID: 40323335 PMCID: PMC12052642 DOI: 10.1007/s12672-025-02471-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is a critical intracellular signaling pathway that is pivotal in various cellular functions. It is in senescence, survival, and growth under normal physiological and pathological conditions, including neoplasms. Additionally, this pathway has been recognized as essential for the regulation of the cell cycle. Several previous studies have indicated that the PI3K/Akt signaling pathway can be influenced by various natural products, with resveratrol (3,4',5-trihydroxy-trans-stilbene) being a particularly important phytoalexin polyphenol in this context. This review explores the impact of the PI3K/Akt signaling pathway on the initiation and advancement of various cancerous conditions and the potential of resveratrol to target this signaling mechanism. The review begins by summarizing the anti-tumor capabilities of resveratrol and then emphasizes the significant role of the PI3K/Akt signaling pathway in the progression of multiple malignancies. Finally, we discuss the therapeutic effects of resveratrol on human neoplasms, from brain cancers to gastrointestinal malignancies, through regulation of this signaling cascade.
Collapse
Affiliation(s)
- Neda Hedayati
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohamad Hosein Safari
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Yaser Eshaghi Milasi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Samaneh Kahkesh
- Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergent Sciences Research Center, TeMs. C., Islamic Azad University, Tehran, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Dorostgou
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergent Sciences Research Center, TeMs. C., Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergent Sciences Research Center, TeMs. C., Islamic Azad University, Tehran, Iran.
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
8
|
Rana M, Liou KC, Thakur A, Nepali K, Liou JP. Advancing glioblastoma therapy: Learning from the past and innovations for the future. Cancer Lett 2025; 617:217601. [PMID: 40037502 DOI: 10.1016/j.canlet.2025.217601] [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: 11/14/2024] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 03/06/2025]
Abstract
Marred by a median survival of only around 12-15 months coupled with poor prognosis and effective therapeutic deprived drug armory, treatment/management of glioblastoma has proved to be a daunting task. Surgical resection, flanked by radiotherapy and chemotherapy with temozolomide, stands as the standard of care; however, this trimodal therapy often manifests limited efficacy due to the heterogeneous and highly infiltrative nature of GBM cells. In addition, the existence of the blood-brain barrier, tumor microenvironment, and the immunosuppressive nature of GBM, along with the encountered resistance of GBM cells towards conventional therapy, also hinders the therapeutic applications of chemotherapeutics in GBM. This review presents key insights into the molecular pathology of GBM, including genetic mutations, signaling pathways, and tumor microenvironment characteristics. Recent innovations such as immunotherapy, oncolytic viral therapies, vaccines, nanotechnology, electric field, and cancer neuroscience, as well as their clinical progress, have been covered. In addition, this compilation also encompasses a discussion on the role of personalized medicine in tailoring treatments based on individual tumor profiles, an approach that is gradually shifting the paradigm in GBM management. Endowed with the learnings imbibed from past failures coupled with the zeal to embrace novel/multidisciplinary approaches, researchers appear to be on the right track to pinpoint more effective and durable solutions in the context of GBM treatment.
Collapse
Affiliation(s)
- Mandeep Rana
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Ke-Chi Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| |
Collapse
|
9
|
Newton F, Halachev M, Nguyen L, McKie L, Mill P, Megaw R. Autophagy disruption and mitochondrial stress precede photoreceptor necroptosis in multiple mouse models of inherited retinal disorders. Nat Commun 2025; 16:4024. [PMID: 40301324 PMCID: PMC12041483 DOI: 10.1038/s41467-025-59165-8] [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: 07/29/2024] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Inherited retinal diseases (IRDs) are a leading cause of blindness worldwide. One of the greatest barriers to developing treatments for IRDs is the heterogeneity of these disorders, with causative mutations identified in over 280 genes. It is therefore a priority to find therapies applicable to a broad range of genetic causes. To do so requires a greater understanding of the common or overlapping molecular pathways that lead to photoreceptor death in IRDs and the molecular processes through which they converge. Here, we characterise the contribution of different cell death mechanisms to photoreceptor degeneration and loss throughout disease progression in humanised mouse models of IRDs. Using single-cell transcriptomics, we identify common transcriptional signatures in degenerating photoreceptors. Further, we show that in genetically and functionally distinct IRD models, common early defects in autophagy and mitochondrial damage exist, triggering photoreceptor cell death by necroptosis in later disease stages. These results suggest that, regardless of the underlying genetic cause, these pathways likely contribute to cell death in IRDs. These insights provide potential therapeutic targets for novel, gene-agnostic treatments for IRDs applicable to the majority of patients.
Collapse
Affiliation(s)
- Fay Newton
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Mihail Halachev
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Linda Nguyen
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Lisa McKie
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Pleasantine Mill
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Roly Megaw
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK.
- Princess Alexandra Eye Pavilion, NHS Lothian, Edinburgh, EH3 9HA, UK.
| |
Collapse
|
10
|
Cao H, Song H, Zhou W, Lv X, Liu X, Xiang Z, Fu R, Cheng Y, Chen J, Wang S, Hu Y, Yan H, You W, Guo C, Chen B, Cao G, Wang W, Jia J. Exploring the active ingredients of Banzhilian and its mechanism of action on diabetic Gastric cancer based on network pharmacology. Sci Rep 2025; 15:14808. [PMID: 40295613 PMCID: PMC12037812 DOI: 10.1038/s41598-025-98214-6] [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: 12/07/2024] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
The incidence of Gastric cancer (GC) has shown a sharp upward trend, and patients with GC complicated by diabetes exhibit significantly worse clinical outcomes and prognosis compared to those without diabetes. Traditional Chinese medicine has played a crucial role in the treatment of both GC and diabetes. Currently, Banzhilian(Scutellaria barbata D. Don) is utilized in the treatment of GC; however, the specific small-molecule monomers it contains and their mechanisms of action have not yet been fully elucidated. This study aims to explore the mechanism of quercetin, a key component of Banzhilian, through network pharmacology, molecular docking, molecular dynamics (MD) simulation, bioinformatics, and in vitro and in vivo experiments. Initially, core targets and key pathways involved in the treatment of diabetes-associated GC (GC-diabetes) were identified using public databases. Subsequently, molecular docking, MD simulation, and survival analysis were performed. Experimental validation included CCK-8 assays, colony formation assays, apoptosis detection, cell cycle analysis, wound healing assays, Transwell migration assays, Western blotting, and mouse subcutaneous tumor formation experiments to evaluate the effects of quercetin, as an active monomer in Banzhilian, on Gastric cancer cells (HGC-27-HG cells) under high-glucose conditions. In this study, quercetin was identified as the key active component, with AKT1, TP53, JUN, MYC, and CCND1 recognized as the target genes, and the PI3K/AKT signaling pathway as the primary regulatory pathway. The results of the study indicate that the proliferation, migration, and invasion capabilities of HGC-27-HG cells are significantly higher than those of HGC-27 cells. However, quercetin inhibited the growth of HGC-27-HG cells, promoted apoptosis, induced cell cycle arrest at the G0/G1 phase, and reduced the cells' migration and invasion abilities. Furthermore, it downregulated the expression of target genes and their phosphorylation levels. The experimental findings confirmed that quercetin, as an active monomer in Banzhilian, suppresses the proliferation of HGC-27-HG cells by inhibiting the PI3K/AKT/MYC pathway, promoting apoptosis, blocking cell cycle progression, and inhibiting cell migration and invasion.
Collapse
Affiliation(s)
- Haikun Cao
- Department of Surgical Oncology, First Affiliated Hospital of Bengbu Medical University, 801 Zhihuai Road, Longzihu District, Bengbu City, 233000, Anhui Province, People's Republic of China
| | - Hui Song
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China
| | - Weiguo Zhou
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei City, 230022, Anhui Province, People's Republic of China
| | - Xiaohu Lv
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China
| | - Xinlei Liu
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China
| | - Zheng Xiang
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China
| | - Rui Fu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei City, 230022, Anhui Province, People's Republic of China
| | - Yixian Cheng
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei City, 230022, Anhui Province, People's Republic of China
| | - Junjie Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei City, 230022, Anhui Province, People's Republic of China
| | - Shengwei Wang
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China
| | - Yvbo Hu
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China
| | - Huayue Yan
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China
| | - Wenlong You
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China
| | - Changqian Guo
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China
| | - Bo Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei City, 230022, Anhui Province, People's Republic of China.
| | - Guodong Cao
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei City, 230022, Anhui Province, People's Republic of China.
| | - Wei Wang
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China.
| | - Jianguang Jia
- Department of Surgical Oncology, First Affiliated Hospital of Bengbu Medical University, 801 Zhihuai Road, Longzihu District, Bengbu City, 233000, Anhui Province, People's Republic of China.
- Department of Surgical Oncology, Second Affiliated Hospital of Bengbu Medical University, No. 633, Longhua Road, Huaishang District, Bengbu City, 233000, Anhui Province, People's Republic of China.
| |
Collapse
|
11
|
Cutano V, Chia ML, Wigmore EM, Hopcroft L, Williamson SC, Christie AL, Willis B, Kerr J, Ashforth J, Fox R, D'Arcy S, Bradshaw L, Blaker C, Eberlein C, Montava-Garriga L, de Bruin EC, Critchlow SE, Brindle KM, Barry ST, Ros S. The interplay between FOXO3 and FOXM1 influences sensitivity to AKT inhibition in PIK3CA and PIK3CA/PTEN altered estrogen receptor positive breast cancer. NPJ Breast Cancer 2025; 11:36. [PMID: 40263319 PMCID: PMC12015352 DOI: 10.1038/s41523-025-00752-9] [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: 09/17/2024] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
Loss of PTEN expression, via homozygous or hemizygous deletion, is common in PIK3CA mutant ER + BC tumors. We assessed reduction of PTEN protein expression on AKT inhibitor capivasertib efficacy in PIK3CA altered tumors. In PIK3CA altered, PTEN protein high models, PI3Kα and AKT inhibition was effective, however ablation and partial PTEN expression reduction attenuated PI3Kαi but not AKTi efficacy, alone or combined with fulvestrant. Efficacy was FOXO3 dependent and associated with FOXM1 downregulation. FOXO3A deletion reduced response to capivasertib, and increased FOXM1 expression. Long term capivasertib exposure of ER+ BC cells upregulated FOXM1 expression. Downregulating FOXM1 expression reversed resistance to capivasertib, while FOXM1 overexpression reduced capivasertib efficacy. Collectively this suggests the AKT-FOXO3-FOXM1 axis plays a pivotal role in response to AKTi in ER+ breast cancer with PIK3CA mutations with and without expression of PTEN, that FOXO3 expression loss can mediate resistance, and that FOXM1 downregulation is a potential biomarker of response.
Collapse
Affiliation(s)
| | - Ming Li Chia
- Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Eleanor M Wigmore
- Early Data Science, Oncology Data Science, AstraZeneca, Cambridge, UK
| | | | | | | | - Brandon Willis
- Bioscience, Early Oncology, AstraZeneca, Boston, MA, USA
| | - James Kerr
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | | | - Rhys Fox
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Sophie D'Arcy
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | | | | | - Cath Eberlein
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | | | | | | | | | - Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Susana Ros
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK.
| |
Collapse
|
12
|
Mulpuri N, Yao XQ, Hamelberg D. Uncovering the Role of Distal Regions in PDK1 Allosteric Activation. ACS BIO & MED CHEM AU 2025; 5:299-309. [PMID: 40255282 PMCID: PMC12006859 DOI: 10.1021/acsbiomedchemau.5c00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 04/22/2025]
Abstract
Allosteric regulation is a pivotal mechanism governing a wide array of cellular functions. Essential to this process is a flexible biomolecule allowing distant sites to interact through coordinated or sequential conformational shifts. Phosphoinositide-dependent kinase 1 (PDK1) possesses a conserved allosteric binding site, the PIF-pocket, which regulates the kinase's ATP binding, catalytic activity, and substrate interactions. We elucidated the allosteric mechanisms of PDK1 by comparing conformational ensembles of the kinase bound with different small-molecule allosteric modulators in the PIF-pocket with that of the modulator-free kinase. Analysis of over 48 μs of simulations consistently shows that the allosteric modulators predominantly influence the conformational dynamics of specific distal regions from the PIF-pocket, driving allosteric activation. Furthermore, a recently developed advanced difference contact network community analysis is employed to elucidate allosteric communications. This approach integrates multiple conformational ensembles into a single community network, offering a valuable tool for future studies aimed at identifying function-related dynamics in proteins.
Collapse
Affiliation(s)
- Nagaraju Mulpuri
- Department
of Chemistry, Georgia State University, Atlanta, Georgia 30302-3965, United
States
| | - Xin-Qiu Yao
- Department
of Chemistry, University of Nebraska at
Omaha, Omaha, Nebraska 68182-0266, United States
| | - Donald Hamelberg
- Department
of Chemistry, Georgia State University, Atlanta, Georgia 30302-3965, United
States
| |
Collapse
|
13
|
Hong Y, He J, Deng D, Liu Q, Zu X, Shen Y. Targeting kinases that regulate programmed cell death: a new therapeutic strategy for breast cancer. J Transl Med 2025; 23:439. [PMID: 40229646 PMCID: PMC11995514 DOI: 10.1186/s12967-025-06367-9] [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: 12/09/2024] [Accepted: 03/08/2025] [Indexed: 04/16/2025] Open
Abstract
Breast cancer is one of the most prevalent malignant tumors among women and ranks as the second leading cause of cancer-related deaths in females, primarily due to delays in diagnosis and shortcomings in treatment strategies. Consequently, there is a pressing need to identify reliable therapeutic targets and strategies. In recent years, the identification of effective biomarkers-particularly novel molecular therapeutic targets-has become a focal point in breast cancer research, aimed at predicting disease aggressiveness and monitoring treatment responses. Simultaneously, advancements in understanding the molecular mechanisms underlying cellular programmed death have opened new avenues for targeting kinase-regulated programmed cell death as a viable therapeutic strategy. This review summarizes the latest research progress regarding kinase-regulated programmed death (including apoptosis, pyroptosis, autophagy, necroptosis, and ferroptosis) in breast cancer treatment. It covers the key kinases involved in this mechanism, their roles in the onset and progression of breast cancer, and strategies for modulating these kinases through pharmacological interventions.
Collapse
Affiliation(s)
- Yun Hong
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-Incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, China
| | - Dan Deng
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-Incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qinyue Liu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-Incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
- Hunan Provincial Clinical Medical Research Center for Drug Evaluation of major chronic diseases, Hengyang, China.
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
- Hunan Provincial Clinical Medical Research Center for Drug Evaluation of major chronic diseases, Hengyang, China.
| |
Collapse
|
14
|
Duarte-Silva AT, Domith I, Gonçalves-da-Silva I, Paes-de-Carvalho R. Vitamin C Modulates the PI3K/AKT Pathway via Glutamate and Nitric Oxide in Developing Avian Retina Cells in Culture. Brain Sci 2025; 15:369. [PMID: 40309873 PMCID: PMC12025763 DOI: 10.3390/brainsci15040369] [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: 03/02/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Background: In addition to its known antioxidant function, the reduced form of vitamin C, ascorbate, also acts as a neuromodulator in the nervous system. Previous work showed a reciprocal interaction of ascorbate with glutamate in chicken embryo retinal cultures. Ascorbate modulates extracellular glutamate levels by inhibiting excitatory amino acid transporter 3 and promoting the activation of NMDA receptors and the consequent activation of intracellular signaling pathways involved in transcription and survival. Objective: In the present work, we investigated the regulation of AKT phosphorylation by ascorbate in chicken embryo retina cultures. Methodology: Cultures of chicken embryo retina cells were tested using Western blot, immunocytochemistry, fluorescent probe transfection, and cellular imaging techniques. Results: Our results show that ascorbate induces a concentration and time-dependent increase in AKT phosphorylation via the accumulation of extracellular glutamate, the activation of glutamate receptors, and the activation of the PI3K pathway. Ascorbate produces an increase in intracellular calcium accumulation and, accordingly, AKT phosphorylation by ascorbate is blocked by the calcium chelator BAPTA-AM. Moreover, AKT phosphorylation is also blocked by the nitric oxide synthase inhibitor 7-nitroindazole, indicating that it is mediated by calcium and nitric oxide-dependent mechanisms. Conclusions: We demonstrate that ascorbate modulates the PI3K/AKT pathway in retinal cultures through the activation of glutamate receptors and NO production in a calcium-dependent manner. Given that previous research has shown that glutamate induces ascorbate release in retinal cultures, our findings emphasize the significance of the reciprocal interactions between ascorbate and glutamate in retinal development. These findings provide further evidence supporting the role of ascorbate as a neuromodulator in retinal development.
Collapse
Affiliation(s)
- Aline T. Duarte-Silva
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24210-346, RJ, Brazil; (A.T.D.-S.); (I.D.); (I.G.-d.-S.)
- Instituto D’Or de Pesquisa e Ensino (IDOR), Rio de Janeiro 22281-100, RJ, Brazil
| | - Ivan Domith
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24210-346, RJ, Brazil; (A.T.D.-S.); (I.D.); (I.G.-d.-S.)
- Instituto D’Or de Pesquisa e Ensino (IDOR), Rio de Janeiro 22281-100, RJ, Brazil
- IDOR/Pioneer Science Initiative, Rio de Janeiro 22281-100, RJ, Brazil
| | - Isabele Gonçalves-da-Silva
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24210-346, RJ, Brazil; (A.T.D.-S.); (I.D.); (I.G.-d.-S.)
| | - Roberto Paes-de-Carvalho
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24210-346, RJ, Brazil; (A.T.D.-S.); (I.D.); (I.G.-d.-S.)
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói 24210-346, RJ, Brazil
| |
Collapse
|
15
|
Konstantaraki M, Berdiaki A, Neagu M, Zurac S, Krasagakis K, Nikitovic D. Understanding Merkel Cell Carcinoma: Pathogenic Signaling, Extracellular Matrix Dynamics, and Novel Treatment Approaches. Cancers (Basel) 2025; 17:1212. [PMID: 40227764 PMCID: PMC11987840 DOI: 10.3390/cancers17071212] [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: 03/01/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Merkel cell carcinoma (MCC) is a rare but aggressive neuroendocrine skin cancer, driven by either Merkel cell polyomavirus (MCPyV) integration or ultraviolet (UV)-induced mutations. In MCPyV-positive tumors, viral T antigens inactivate tumor suppressors pRb and p53, while virus-negative MCCs harbor UV-induced mutations that activate similar oncogenic pathways. Key signaling cascades, including PI3K/AKT/mTOR and MAPK, support tumor proliferation, survival, and resistance to apoptosis. Histologically, MCC consists of small round blue cells with neuroendocrine features, high mitotic rate, and necrosis. The tumor microenvironment (TME) plays a central role in disease progression and immune escape. It comprises a mix of tumor-associated macrophages, regulatory and cytotoxic T cells, and elevated expression of immune checkpoint molecules such as PD-L1, contributing to an immunosuppressive niche. The extracellular matrix (ECM) within the TME is rich in proteoglycans, collagens, and matrix metalloproteinases (MMPs), facilitating tumor cell adhesion, invasion, and interaction with stromal and immune cells. ECM remodeling and integrin-mediated signaling further promote immune evasion and therapy resistance. Although immune checkpoint inhibitors targeting PD-1/PD-L1 have shown promise in treating MCC, resistance remains a major hurdle. Therapeutic strategies that concurrently target the TME-through inhibition of ECM components, MMPs, or integrin signaling-may enhance immune responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Maria Konstantaraki
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
- Dermatology Department, University Hospital of Heraklion, 71110 Heraklion, Greece;
| | - Aikaterini Berdiaki
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
| | - Monica Neagu
- Immunology Laboratory, “Victor Babes” National Institute of Pathology, 99-101 Splaiul Independenței, 050096 Bucharest, Romania;
- Pathology Department, Colentina Clinical Hospital, 19-21 Sos Stefan Cel Mare, 020125 Bucharest, Romania;
| | - Sabina Zurac
- Pathology Department, Colentina Clinical Hospital, 19-21 Sos Stefan Cel Mare, 020125 Bucharest, Romania;
- Faculty of Dentistry, University of Medicine and Pharmacy, 8 Eroilor Sanitari Boulevard, 050474 Bucharest, Romania
| | | | - Dragana Nikitovic
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
| |
Collapse
|
16
|
Pan Z, Liu Y, Li H, Qiu H, Zhang P, Li Z, Wang X, Tian Y, Feng Z, Zhu S, Wang X. The role and mechanism of aerobic glycolysis in nasopharyngeal carcinoma. PeerJ 2025; 13:e19213. [PMID: 40191756 PMCID: PMC11971989 DOI: 10.7717/peerj.19213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
This review delves into the pivotal role and intricate mechanisms of aerobic glycolysis in nasopharyngeal carcinoma (NPC). NPC, a malignancy originating from the nasopharyngeal epithelium, displays distinct geographical and clinical features. The article emphasizes the significance of aerobic glycolysis, a pivotal metabolic alteration in cancer cells, in NPC progression. Key enzymes such as hexokinase 2, lactate dehydrogenase A, phosphofructokinase 1, and pyruvate kinase M2 are discussed for their regulatory functions in NPC glycolysis through signaling pathways like PI3K/Akt and mTOR. Further, the article explores how oncogenic signaling pathways and transcription factors like c-Myc and HIF-1α modulate aerobic glycolysis, thereby affecting NPC's proliferation, invasion, metastasis, angiogenesis, and immune evasion. By elucidating these mechanisms, the review aims to advance research and clinical practice in NPC, informing the development of targeted therapeutic strategies that enhance treatment precision and reduce side effects. Overall, this review offers a broad understanding of the multifaceted role of aerobic glycolysis in NPC and its potential impact on therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiyong Pan
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuyi Liu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Hui Li
- Department of Ophthalmology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Huisi Qiu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Pingmei Zhang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhiying Li
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xinyu Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuxiao Tian
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhengfu Feng
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Song Zhu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xin Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| |
Collapse
|
17
|
Liao S, Zhang X, Chen L, Zhang J, Lu W, Rao M, Zhang Y, Ye Z, Ivanova D, Li F, Chen X, Wang Y, Song A, Xie B, Wang M. KRT14 is a promising prognostic biomarker of breast cancer related to immune infiltration. Mol Immunol 2025; 180:55-73. [PMID: 40014952 DOI: 10.1016/j.molimm.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/12/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Breast cancer (BC) is the leading cancer among women globally, which has the highest incidence and mortality rate in over a hundred countries. This study was intended to discover a new prognostic biomarker, facilitating personalized treatment approaches. METHODS RNA sequencing data from The Cancer Genome Atlas database and Gene Expression Omnibus database were utilized to download to evaluate expression levels and prognostic significance of Keratin 14 (KRT14). Methylation of KRT14 was also assessed. The CIBERSORT and single-sample gene set enrichment analysis algorithms were applied to explore the connection between KRT14 and the tumor microenvironment. Primary drugs' sensitivity and potential small molecule therapeutic compounds were analyzed through the "pRRophetic" R package and the Connectivity Map. The prognostic value of KRT14 was additionally corroborated through a comparison of protein levels in peritumoral and cancerous tissues via immunohistochemistry. Moreover, an immune-related prognostic model based on KRT14 was designed to enhance the prediction accuracy for the prognosis of BC patients. RESULTS The study found that KRT14 expression was generally downregulated in BC, correlating strongly with poor prognosis. Compared to normal tissues, the methylation level of KRT14 was higher in BC tissues. Lower expression of KRT14 was linked to decreased anti-tumoral immune cells infiltration and increased immunosuppressive cells infiltration. Sensitivity to various key therapeutic drugs was lower in groups with diminished KRT14 expression. In addition, several potential anti-BC small molecule compounds were identified. The model designed in this study significantly enhanced the predictive capability for BC patients compared to predictions based solely on KRT14 expression levels. CONCLUSION Overall, KRT14 was closely correlated with the prognosis in BC, making it a reliable biomarker.
Collapse
Affiliation(s)
- Siqi Liao
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xin Zhang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lanhui Chen
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jianning Zhang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Weiyu Lu
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Mengou Rao
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yifan Zhang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zijian Ye
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Deyana Ivanova
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston MA02115, USA
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Xuemei Chen
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Anchao Song
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Biao Xie
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| | - Meijiao Wang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China; Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
18
|
Ding Z, Shao G, Li M. Targeting autophagy in premature ovarian failure: Therapeutic strategies from molecular pathways to clinical applications. Life Sci 2025; 366-367:123473. [PMID: 39971127 DOI: 10.1016/j.lfs.2025.123473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Premature ovarian failure (POF) is a condition where the ovaries lose their function before the age of 40, leading to significant impacts on reproductive health and overall well-being. Current treatment options are limited and often ineffective at restoring ovarian function. This review explores the role of autophagy- a cellular process that helps maintain homeostasis by recycling damaged components-in the development and potential treatment of POF. Autophagy is crucial for the survival of follicle cells and can be disrupted by various stressors associated with POF, such as oxidative damage and mitochondrial dysfunction. We review several key molecular pathways involved in autophagy, including the PI3K/AKT/mTOR, PINK1-Parkin, JAK2/STAT3, MAPK and AMPK/FOXO3a pathways, which have been implicated in POF. Each pathway offers unique insights into how autophagy can be modulated to counteract POF-related damage. Additionally, we discuss emerging therapeutic strategies that target these pathways, including chemical compounds, peptides, hormones, RNA therapy, extracellular vesicles and traditional Chinese medicine. These approaches aim to restore autophagic balance, promote follicle survival and improve ovarian function. By targeting autophagy, new treatments may offer hope for better management and potential reversal of POF, thus improving the quality of life for affected individuals.
Collapse
Affiliation(s)
- Ziwen Ding
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Genbao Shao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Mingyang Li
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
19
|
Li C, Cui J, Zheng H, Sha Z, Wei R, Wu R, Ni B. The NADC30-like PRRSV activates the integrin αV subunit to facilitate its entry into Marc-145 cells. PLoS One 2025; 20:e0316239. [PMID: 40146709 PMCID: PMC11949365 DOI: 10.1371/journal.pone.0316239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/08/2024] [Indexed: 03/29/2025] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a highly contagious virus that poses a significant threat to the global pig farming industry, resulting in substantial economic losses. However, owing to the high variability of PRRSV and unclear mechanisms of infection, there are currently no effective vaccines or drugs available for its prevention and control. Our previous report revealed that highly pathogenic PRRSV (HP-PRRSV) requires the FAK-PI3K-AKT signaling pathway to facilitate its entry into cells. In this study, we further investigated whether the integrin subunit was involved in the entry process of NADC30-like PRRSV. First, the integrin subunits in Marc-145 cells were characterized by RT-PCR, and 11 of these subunits were identified, nearly all of which interacted with the integrin α V and β1 subunits to form heterodimers. Western blot analysis revealed that the integrin α V subunit was highly expressed in Marc-145 cells, and blocking this subunit with a functional antibody or siRNA significantly attenuated NADC30-like PRRSV entry without affecting virus binding. Moreover, in Marc-145 cells, NADC30-like PRRSV could activate the FAK-PI3K-AKT signaling pathway through the integrin α V subunit. Blocking the α V subunit significantly inhibited signal transduction and virus entry, and treatment of cells with the PI3K activator greatly reversed this inhibitory effect. Furthermore, the α V subunit activator manganese could also enhance NADC30-like PRRSV entry and signal transduction. In conclusion, our results revealed that NADC30-like PRRSV could activate the integrin α V subunit and subsequently transduce signals to the FAK-PI3K-AKT signaling pathway to facilitate entry into Marc-145 cells.
Collapse
Affiliation(s)
- Chunlin Li
- China Animal Health and Epidemiology Center, Qingdao, China
- Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jin Cui
- China Animal Health and Epidemiology Center, Qingdao, China
- Key Laboratory of Animal Biosafety Risk Prevention and Control (South), Ministry of Agriculture and Rural Affairs, Qingdao, People’s Republic of China
- Key Laboratory of Animal Biosafety, Qingdao, China
| | - Hui Zheng
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Zhou Sha
- China Animal Health and Epidemiology Center, Qingdao, China
- Key Laboratory of Animal Biosafety Risk Prevention and Control (South), Ministry of Agriculture and Rural Affairs, Qingdao, People’s Republic of China
- Key Laboratory of Animal Biosafety, Qingdao, China
| | - Rong Wei
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Rui Wu
- Heilongjiang Bayi Agricultural University, Daqing, China
- Jiamusi University, Jiamusi, China
| | - Bo Ni
- China Animal Health and Epidemiology Center, Qingdao, China
- Key Laboratory of Animal Biosafety Risk Prevention and Control (South), Ministry of Agriculture and Rural Affairs, Qingdao, People’s Republic of China
- Key Laboratory of Animal Biosafety, Qingdao, China
- Qingdao Key Laboratory of Modern Bioengineering and Animal Disease Research, Qingdao, China
| |
Collapse
|
20
|
Torosyan H, Paul MD, Maker A, Meyer BG, Jura N, Verba KA. Structures of the PI3Kα/KRas complex on lipid bilayers reveal the molecular mechanism of PI3Kα activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.22.644753. [PMID: 40196507 PMCID: PMC11974675 DOI: 10.1101/2025.03.22.644753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
PI3Kα is a potent oncogene that converts PIP2 to PIP3 at the plasma membrane upon activation by receptor tyrosine kinases and Ras GTPases. In the absence of any structures of activated PI3Kα, the molecular details of its activation remain unknown. Here, we present cryo-EM structures of the PI3Kα/KRas complex embedded in lipid nanodiscs, revealing a rich ensemble of PI3Kα states adopted at the membrane surface. The sequential addition of a lipid bilayer, PIP2 and an activating phosphopeptide leads to the progressive release of key inhibitory domains from the PI3Kα catalytic core, which directly correlates with the reorganization of its active site. While association with POPC/POPS nanodiscs partially relieves PI3Kα autoinhibition, incorporation of PIP2 triggers near-complete displacement of PI3Kα inhibitory domains and significant restructuring of active site regulatory motifs. The addition of the activating phosphopeptide induces dimerization of the PI3Kα/KRas complex through a p110α catalytic subunit-mediated interface that is sterically occluded in autoinhibited PI3Kα. In cells, this dimeric PI3Kα complex amplifies Akt signaling in response to growth factor stimulation. Collectively, our structures map the conformational landscape of PI3Kα activation and reveal previously unexplored interfaces for potential therapeutic targeting.
Collapse
|
21
|
Bao ZC, Liu ZD, Zhang Y, Dai HJ, Jia H, Ren F, Li N, Zhao L, Wang YW, Lv SY, Zhang Y. To investigate the effect and mechanism of tetrahydrocurcumin on hepatocellular carcinoma based on phosphoinositide 3-kinases/AKT signaling pathway. World J Gastrointest Oncol 2025; 17:102187. [PMID: 40092949 PMCID: PMC11866248 DOI: 10.4251/wjgo.v17.i3.102187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/20/2024] [Accepted: 01/02/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Liver cancer has a high incidence and mortality worldwide, especially in China. Herein, we investigated the therapeutic effect and mechanism of tetrahydrocurcumin against hepatocellular carcinoma (HCC), with a focus on the of phosphoinositide 3-kinases (PI3K)/AKT signaling pathway. AIM To investigate the effects and mechanism of tetrahydrocurcumin in HCC cell lines HepG2 and Huh7. METHODS Using Metascape, we analyzed the potential targets of tetrahydrocurcumin in HCC. Molecular docking validation was performed using SYBYL2.0. Cell Counting Kit-8, wound healing, and transwell assays were performed to evaluate the effects of tetrahydrocurcumin on HepG2 and Huh7 cell migration, invasion, and apoptosis. The expression of PI3K/AKT signaling pathway-related proteins was detected by western blotting. RESULTS Network pharmacology and molecular docking showed that tetrahydrocurcumin has high binding affinity for phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha. In vitro experiments demonstrated that tetrahydrocurcumin suppressed the migration and invasion of liver cancer cells, promoted their apoptosis, and downregulated the expression of p-PI3K, p-AKT, and B cell leukemia/lymphoma 2, while upregulating caspase-3, p53, and B cell leukemia/lymphoma 2 associated X. CONCLUSION In summary, tetrahydrocurcumin suppresses PI3K/AKT signaling, promotes apoptosis, and prevents the migration and invasion of liver cancer cells.
Collapse
Affiliation(s)
- Zhuo-Cong Bao
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Zhao-Dong Liu
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Ye Zhang
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Hui-Jun Dai
- Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Hui Jia
- School of Traditional Chinese Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Fu Ren
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Ning Li
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Lu Zhao
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Yi-Wei Wang
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Molecular Morphology Laboratory, College of Basic Medical Sciences, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Shang-Yu Lv
- Batch 2022, Clinical Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Yan Zhang
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- International Education School, International Exchange and Cooperation Office, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| |
Collapse
|
22
|
Matsumoto K, Matsumoto Y, Wada J. PARylation-mediated post-transcriptional modifications in cancer immunity and immunotherapy. Front Immunol 2025; 16:1537615. [PMID: 40134437 PMCID: PMC11933034 DOI: 10.3389/fimmu.2025.1537615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Poly-ADP-ribosylation (PARylation) is a post-translational modification in which ADP-ribose is added to substrate proteins. PARylation is mediated by a superfamily of ADP-ribosyl transferases known as PARPs and influences a wide range of cellular functions, including genome integrity maintenance, and the regulation of proliferation and differentiation. We and others have recently reported that PARylation of SH3 domain-binding protein 2 (3BP2) plays a role in bone metabolism, immune system regulation, and cytokine production. Additionally, PARylation has recently gained attention as a target for cancer treatment. In this review, we provide an overview of PARylation, its involvement in several signaling pathways related to cancer immunity, and the potential of combination therapies with PARP inhibitors and immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Yoshinori Matsumoto
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Faculty of
Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | |
Collapse
|
23
|
Longfei H, Wenyuan H, Weihua F, Peng P, Sun L, Kun L, Mincong H, Fan Y, Wei H, Qiushi W. Exosomes in cartilage microenvironment regulation and cartilage repair. Front Cell Dev Biol 2025; 13:1460416. [PMID: 40109360 PMCID: PMC11919854 DOI: 10.3389/fcell.2025.1460416] [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: 07/06/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Osteoarthritis (OA) is a debilitating disease that predominantly impacts the hip, hand, and knee joints. Its pathology is defined by the progressive degradation of articular cartilage, formation of bone spurs, and synovial inflammation, resulting in pain, joint function limitations, and substantial societal and familial burdens. Current treatment strategies primarily target pain alleviation, yet improved interventions addressing the underlying disease pathology are scarce. Recently, exosomes have emerged as a subject of growing interest in OA therapy. Numerous studies have investigated exosomes to offer promising therapeutic approaches for OA through diverse in vivo and in vitro models, elucidating the mechanisms by which exosomes from various cell sources modulate the cartilage microenvironment and promote cartilage repair. Preclinical investigations have demonstrated the regulatory effects of exosomes originating from human cells, including mesenchymal stem cells (MSC), synovial fibroblasts, chondrocytes, macrophages, and exosomes derived from Chinese herbal medicines, on the modulation of the cartilage microenvironment and cartilage repair through diverse signaling pathways. Additionally, therapeutic mechanisms encompass cartilage inflammation, degradation of the cartilage matrix, proliferation and migration of chondrocytes, autophagy, apoptosis, and mitigation of oxidative stress. An increasing number of exosome carrier scaffolds are under development. Our review adopts a multidimensional approach to enhance comprehension of the pivotal therapeutic functions exerted by exosomes sourced from diverse cell types in OA. Ultimately, our aim is to pinpoint therapeutic targets capable of regulating the cartilage microenvironment and facilitating cartilage repair in OA.
Collapse
Affiliation(s)
- Han Longfei
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hou Wenyuan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Fang Weihua
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Peng Peng
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lu Sun
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lin Kun
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Mincong
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yang Fan
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Wei
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei Qiushi
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Orthopaedic, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
24
|
Song BF, Li BJ, Sun Y, Li M, Rao T, Ruan Y, Cheng F. GOLPH3 promotes calcium oxalate-induced renal injury and fibrosis through Golgi stress-mediated apoptosis and inflammatory responses. Sci Rep 2025; 15:7640. [PMID: 40038402 PMCID: PMC11880244 DOI: 10.1038/s41598-025-91638-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
A common urological disorder, calcium oxalate (CaOx) stones are the most common form of kidney stones. Deposition of CaOx crystals leads to tubular damage, interstitial fibrosis, and chronic kidney disease. Understanding the intrinsic mechanisms of kidney stone formation is essential for the prevention of kidney stones and the development of new therapeutic agents. The Golgi apparatus is a key organelle in the secretory pathway of eukaryotic cells, which plays an important role in the sorting, modification, and transport of proteins within the cell, and has been reported to be involved in several diseases, including prostate tumors, gastrointestinal tumors, sepsis, and so on. GOLPH3 is also known as GPP34, GMx33, or MIDAS. It is a glycoprotein that regulates traffic between the trans-Golgi network and the cell membrane. However, its role in renal injury caused by CaOx crystal deposition is still unclear. Results from immunohistochemistry, qRT-PCR, western blot, and public database single nucleotide RNA-seq showed that GOLPH3 was significantly upregulated in kidney stone patients and animal kidneys. Significant inhibition of Golgi stress, apoptosis, and renal fibrosis by GOLPH3 inhibition with siRNA in CaOx-stimulated HK-2 cells. The PI3K\AKT\mTOR signaling pathway was inhibited by GOLPH3 knockdown, which may be associated with reduced inflammatory response and apoptosis, as well as restoration of Golgi morphology and function. In conclusion, GOLPH3 plays a critical role in CaOx-induced kidney injury by promoting Golgi stress and increasing inflammatory responses, apoptosis, and renal fibrosis, suggesting that GOLPH3 is a potential therapeutic target for kidney stones.
Collapse
Affiliation(s)
- Bao-Feng Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bo-Jun Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yushi Sun
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ming Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yuan Ruan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
25
|
Wen L, Rong F, Dai G, Liu Y, Lv Y, Luo Q, Liu DX, Chen R. Proteomic analysis of the nonstructural protein 2-host protein interactome reveals a novel regulatory role of SH3 domain-containing kinase-binding protein 1 in porcine reproductive and respiratory syndrome virus replication and apoptosis. Int J Biol Macromol 2025; 295:139218. [PMID: 39755310 DOI: 10.1016/j.ijbiomac.2024.139218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025]
Abstract
Virus-host protein interaction is critical for successful completion of viral replication cycles. As the largest nonstructural protein (NSP) of porcine reproductive and respiratory syndrome virus (PRRSV), NSP2 plays multiple and critical roles in viral replication, antiviral immunity, cellular tropism and virulence. An interactome of this protein with host proteins would be instrumental in full understanding of these multifunctional roles. In this study, we report the identification of 120 NSP2-interacting host proteins by co-immunoprecipitation coupled liquid chromatography mass spectrometry, via rescuing and utilizing a recombinant PRRSV expressing an HA-tagged NSP2. By comparing and subtracting with cells infected with parental virus, a comprehensive interactome was constructed. Bioinformatics analysis revealed that these host factors are mainly involved in translation regulation, metabolism, signal transduction and innate immunity signaling pathways. Selection of five host proteins (CtBP1, CtBP2, HSPA2, PPP1CA, SH3KBP1) for further verification and characterization confirmed their interactions with NSP2 and differential effects on PRRSV replication. Intriguingly, interaction of NSP2 and SH3KBP1 led to specific cleavage of SH3KBP1, antagonizing its pro-apoptotic activity. Taken together, this study provides overarching views on the NSP2-host interactome, paving a solid foundation for functional studies of host proteins in PRRSV biology and their potential as targets for novel therapeutics development.
Collapse
Affiliation(s)
- Lianghai Wen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China; Zhaoqing Institute of Biotechnology Co., Ltd., Zhaoqing 526238, China
| | - Fang Rong
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| | - Guo Dai
- Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China
| | - Yufu Liu
- School of Life Sciences, Zhaoqing University, Zhaoqing 526061, China
| | - Yadi Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| | - Qiong Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Wens Dahuanong Bio-Pharmaceutical Co., Ltd., Xinxing 527400, China
| | - Ding Xiang Liu
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China; Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China.
| | - Ruiai Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China; Zhaoqing Institute of Biotechnology Co., Ltd., Zhaoqing 526238, China; Guangdong Wens Dahuanong Bio-Pharmaceutical Co., Ltd., Xinxing 527400, China.
| |
Collapse
|
26
|
Wang P, Mak VC, Rao L, Wu Q, Zhou Y, Sharma R, Kwon SC, Cheung LW. p85β acts as a transcription cofactor and cooperates with BCLAF1 in the nucleus. Nat Commun 2025; 16:2042. [PMID: 40016211 PMCID: PMC11868507 DOI: 10.1038/s41467-025-56532-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/16/2025] [Indexed: 03/01/2025] Open
Abstract
p85β is a regulatory subunit of the phosphoinositide 3-kinase (PI3K). Emerging evidence suggests that p85β goes beyond its role in the PI3K and is functional in the nucleus. In this study, we discover that nuclear p85β is enriched at gene loci and regulates gene transcription and that this regulatory role contributes to the oncogenic potential of nuclear p85β. A multi-omics approach reveals the physical interaction and functional cooperativity between nuclear p85β and a transcription factor BCLAF1. We observe genome-wide co-occupancy of p85β and BCLAF1 at gene targets associated with transcriptional responses. Intriguingly, the targetome includes BCLAF1 of which transcription is activated by p85β and BCLAF1, indicating a positive autoregulation. While BCLAF1 recruits p85β to BCLAF1 loci, p85β facilitates the assembly of BCLAF1, the scaffold protein TRIM28 and the zinc finger transcription factor ZNF263, which together act in concert to activate BCLAF1 transcription. Collectively, this study provides functional evidence and mechanistic basis to support a role of nuclear p85β in modulating gene transcription.
Collapse
Affiliation(s)
- Panpan Wang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Victor Cy Mak
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ling Rao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qiuqiu Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuan Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Rakesh Sharma
- Proteomics and Metabolomics Core, Centre for PanorOmic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - S Chul Kwon
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lydia Wt Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
27
|
Zhang HL, Lin Z, Zhang Y. Developments in research and commercialization of PI3K and AKT targets: a patent-based landscape. Pharm Pat Anal 2025:1-8. [PMID: 39993965 DOI: 10.1080/20468954.2025.2470102] [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: 10/11/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025]
Abstract
PI3K and AKT signaling pathway has been linked to the pathophysiology of various diseases. This pathway has emerged as a crucial therapeutic strategy for cancer and other diseases. To better understand recent development of PI3K and AKT, a patent-based landscape study was performed. The results shows that both PI3K and AKT targets have shown prolific patent filings over the past 20 years. This study is the first to depict the therapeutic applications of both PI3K and AKT targets based on a patent big data analysis. Ten key therapeutic applications were identified, with over 77% of patents related to anti-cancer therapy for both PI3K and AKT targets. Additionally, our findings show that combination therapy is a distinguishing feature for drugs targeting both PI3K and AKT. The average time from patent application to drug approval for PI3K target drugs is 8.8 years. PI3K target drugs obtain market approval faster compared to AKT drugs. Approximately, 2 years of patent term extension could be obtained if the time from the patent application date to the drug approval date is less than 10 years.
Collapse
Affiliation(s)
- Hai-Long Zhang
- Central International Intellectual Property (Baotou) Co. Ltd, Baotou, China
| | - Zhaochen Lin
- Hydrogen Medicine Research Centre, The Affiliated Taian City Central Hospital of Qingdao University, Tai'an, Shandong, China
| | - Ying Zhang
- Pharmacy Intravenous Admixture Services, The Affiliated Taian City Central Hospital of Qingdao University, Tai'an, Shandong, China
| |
Collapse
|
28
|
Sellitto C, White TW. Combinatorial genetic manipulation of Cx50, PI3K and PTEN alters postnatal mouse lens growth and homeostasis. FRONTIERS IN OPHTHALMOLOGY 2025; 5:1502836. [PMID: 40046897 PMCID: PMC11879993 DOI: 10.3389/fopht.2025.1502836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025]
Abstract
Introduction Phosphoinositide 3-kinase (PI3K), Phosphatase and tensin homolog (PTEN) and connexin50 (Cx50) have individually been shown to play critical roles in the growth, development and maintenance of the lens and to functionally interact in vitro. To elucidate how gap junctional coupling mediated by Cx50 and intracellular signaling mediated by PI3K and PTEN synergistically interact to regulate lens homeostasis in vivo, we generated and characterized double knockout animal models lacking the p110α subunit of PI3K and Cx50, or PTEN and Cx50. Methods We interbred lens specific p110α and PTEN conditional knockout animals with Cx50 deficient mice to generate double knockouts. Animals and eyes were weighed, lenses were dissected, photographed, measured, fixed and sectioned for histological analysis. Lens epithelial cell proliferation was determined using 5-ethynyl-2'-deoxyuridine (EdU) labeling. Results Double knockout of p110α and Cx50 led to a significant reduction in lens and eye size, and a high rate of lens rupture. The individual cell proliferation defects of the Cx50 and p110α single knockout lenses both persisted in the double KO. Double deletion of Cx50 and PTEN produced severe lens defects, including cataract, aberrant cell migration, altered cell proliferation, vacuole formation and lens rupture. Conclusion The severe phenotypes in p110α/Cx50 and PTEN/Cx50 double deficient lenses suggest that PI3K, PTEN and Cx50 participate in both distinct and common regulatory pathways that are necessary to maintain normal lens growth and homeostasis.
Collapse
Affiliation(s)
| | - Thomas W. White
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, United States
| |
Collapse
|
29
|
Qiang M, Chen Z, Liu H, Dong J, Gong K, Zhang X, Huo P, Zhu J, Shao Y, Ma J, Zhang B, Liu W, Tang M. Targeting the PI3K/AKT/mTOR pathway in lung cancer: mechanisms and therapeutic targeting. Front Pharmacol 2025; 16:1516583. [PMID: 40041495 PMCID: PMC11877449 DOI: 10.3389/fphar.2025.1516583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
Owing to its high mortality rate, lung cancer (LC) remains the most common cancer worldwide, with the highest malignancy diagnosis rate. The phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling (PAM) pathway is a critical intracellular pathway involved in various cellular functions and regulates numerous cellular processes, including growth, survival, proliferation, metabolism, apoptosis, invasion, and angiogenesis. This review aims to highlight preclinical and clinical studies focusing on the PAM signaling pathway in LC and underscore the potential of natural products targeting it. Additionally, this review synthesizes the existing literature and discusses combination therapy and future directions for LC treatment while acknowledging the ongoing challenges in the field. Continuous development of novel therapeutic agents, technologies, and precision medicine offers an increasingly optimistic outlook for the treatment of LC.
Collapse
Affiliation(s)
- Min Qiang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Zhe Chen
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hongyang Liu
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Junxue Dong
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Kejian Gong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xinjun Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Peng Huo
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jingjun Zhu
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yifeng Shao
- Department of General Surgery, Capital Institute of Pediatrics’ Children’s Hospital, Beijing, China
| | - Jinazun Ma
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Bowei Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Mingbo Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Zeng M, Hu Y, Zhao L, Duan C, Wu H, Xu Y, Liu X, Wang Y, Jiang D, Zeng S. Design, synthesis, and pharmacological evaluation of triazine-based PI3K/mTOR inhibitors for the potential treatment of non-small cell lung cancer. Eur J Med Chem 2025; 284:117200. [PMID: 39733482 DOI: 10.1016/j.ejmech.2024.117200] [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: 10/14/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 12/31/2024]
Abstract
Dysregulated activation of the PI3K/AKT/mTOR pathway is crucial in the development of cancer, and disrupting it could potentially lead to cancer suppression, making it a viable strategy for cancer treatment. Here, as a consecutive work of our team, we described the identification and optimization of PI3K/mTOR inhibitors based on triazine scaffold, which exhibited potent PI3K/mTOR inhibitor activity. The systematically structure-activity relationship (SAR) results demonstrated that compound 5nh displayed high efficacy against PI3Kα and mTOR, with the IC50 values of 0.45 nM and 2.9 nM, respectively. Importantly, compared to the lead compound PKI-587, 5nh demonstrated significant inhibitory activity against non-small-cell lung cancer (NSCLC) cell lines, particularly HCC-827, with a 43-fold increase (3.5 nM vs 150 nM). Additionally, the compound showed effective inhibition against the EGFR-resistant variant HCC-827(GR) cell line. Mechanism validation demonstrated that 5nh significantly interfered with the PI3K/AKT/mTOR signaling pathway in HCC-827 cells. Furthermore, the oral pharmacokinetic properties of 5nh had been observably improved, with AUC0-t and Cmax increasing by 13-16 times at a dose of 10 mg/kg in mice. Importantly, the in vivo efficacy study demonstrated that orally treatment of 5nh led to significant tumor growth suppression, with a TGI value of 84.4 %. Collectively, our systematically medicinal chemistry campaigns suggested that 5nh, a novel oral available triazine derivative, held promise as a candidate for therapy of NSCLC by targeting the PI3K/AKT/mTOR cascade.
Collapse
Affiliation(s)
- Ming Zeng
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China.
| | - Yingxuan Hu
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Lan Zhao
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Chengze Duan
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310053, China
| | - Haifeng Wu
- Zhejiang Research Institute of Chemical Industry, Hangzhou, 310023, China
| | - Yi Xu
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Xiaoguang Liu
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Yali Wang
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Dengzhao Jiang
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Shenxin Zeng
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310053, China; School of Pharmacy, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
31
|
Wei Y, Ma Z, Li Z, Kang J, Liao T, Jie L, Liu D, Shi L, Wang P, Mao J, Wu P. Gentiopicroside ameliorates synovial inflammation and fibrosis in KOA rats by modulating the HMGB1-mediated PI3K/AKT signaling axis. Int Immunopharmacol 2025; 147:113973. [PMID: 39764995 DOI: 10.1016/j.intimp.2024.113973] [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: 10/24/2024] [Revised: 12/17/2024] [Accepted: 12/28/2024] [Indexed: 01/29/2025]
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a degenerative joint disease characterized by synovial inflammation and fibrosis. Gentiopicroside (GPS), one of the main active ingredients of Gentiana macrophylla, is widely used in anti-inflammatory and anti-fibrotic therapies. However, the exact mechanism by which GPS treats synovial inflammation and fibrosis in KOA remains unclear. METHODS Fibroblast-like synoviocytes (FLSs) were stimulated with lipopolysaccharide (LPS) to induce inflammation and fibrosis, and CCK-8 was performed to determine the viability of GPS-treated FLSs, using immunofluorescence to examine the expression of P-PI3K and P-AKT, confocal microscopy was used to identify intracellular HMGB1 translocation. The KOA rat model was established by anterior cruciate ligament transection (ACLT) and subsequently subjected to GPS intervention. Inflammatory cytokines (TNF-α, IL-1β, and IL-6), fibrosis-related indicators (TGF-β, collagen I, TIMP1, and α-SMA), and HMGB1/PI3K/AKT signaling axis-related proteins and gene expression of fibroblast-like synoviocytes and synovial tissues were detected by Western blotting and real-time PCR. The histopathology of the synovium of the rats was assessed using Hematoxylin-eosin (HE), Sirius Red, and Masson staining. Immunohistochemistry was performed to detect the expression of HMGB1, P-PI3K, and P-AKT. RESULTS The present study revealed that GPS intervention significantly ameliorated inflammation and fibrosis in LPS-stimulated FLSs and KOA rat synovium. Immunofluorescence demonstrated that GPS inhibited the release of HMGB1 from the nucleus. Furthermore, GPS intervention down-regulates the levels of proteins and gene associated with the HMGB1/PI3K/AKT signaling pathway. CONCLUSION GPS ameliorated synovial inflammation and fibrosis in KOA rats, which may involve HMGB1-mediated activation of the PI3K/AKT signaling axis.
Collapse
Affiliation(s)
- Yibao Wei
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Zhenyuan Ma
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Zhenhui Li
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Junfeng Kang
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; The Affiliated Hospital of Shanxi University of Traditional Chinese Medicine, Taiyuan 030002, China
| | - Taiyang Liao
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lishi Jie
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Deren Liu
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lei Shi
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Peimin Wang
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Jun Mao
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China.
| | - Peng Wu
- Department of Orthopaedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
32
|
Wei L, Chen P, Shi L, Li G, Feng X, Zhao Y, Wang J, Chen ZS, Hu Z, Cui M, Zhou B. Composite Graphene for the Dimension- and Pore-Size-Mediated Stem Cell Differentiation to Bone Regenerative Medicine. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7307-7323. [PMID: 39843162 DOI: 10.1021/acsami.4c17554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
As one of the most promising means to repair diseased tissues, stem cell therapy with immense potential to differentiate into mature specialized cells has been rapidly developed. However, the clinical application of stem-cell-dominated regenerative medicine was heavily hindered by the loss of pluripotency during the long-term in vitro expansion. Here, a composite three-dimensional (3D) graphene-based biomaterial, denoted as GO-Por-CMP@CaP, with hierarchical pore structure (micro- to macropore), was developed to guide the directional differentiation of human umbilical cord MSCs (hucMSCs) into osteoblasts. GO-Por-CMP@CaP could act as a high-efficiency living composite material without a "dead space", effectively regulating the cellular response. The 3D topological structure generated via the two-step modification on two-dimensional graphene could effectively mimic the natural 3D microenvironment of cells, enhancing the stem cell attachment, which is not only conducive for the proliferation of stem cells but also beneficial for the osteogenic differentiation. Meanwhile, the wide existence of interconnected macropores was favorable for bone ingrowth, capillary formation, as well as the nutrients transportation. Furthermore, the concurrent existence of micro- and mesopores significantly promoted the extracellular matrix (ECM) adsorption, which ensured cellular attachment, leading to multiscale osteointegration. Both in vitro and in vivo assay demonstrated the above three factors collaborated mutually with nanosized calcium phosphate (CaP, with chemical similarities to the inorganic components of bone), which provided abundant adhesive sites to adequately induce osteogenic differentiation in the absence of any soluble growth factors. Proteomic analysis experiments confirmed that GO-Por-CMP@CaP promoted the differentiation of hucMSCs cells into osteoblasts by affecting the PI3K-Akt signaling pathway through the up-regulation of SPP1 protein. Our study offers a pure material-based stem cell differentiation regulating behavior via engineering the dimension and porosity of material, which provides insights into the design and development of substitutes to bone repair materials.
Collapse
Affiliation(s)
- Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong PR China
| | - Peilei Chen
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong PR China
| | - Lin Shi
- Weifang People's Hospital, Shandong Second Medical University, Weifang 261035, Shandong PR China
| | - Gentao Li
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong PR China
| | - Xiaozhe Feng
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong PR China
| | - Yao Zhao
- Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261035, Shandong PR China
| | - Jiangyun Wang
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong PR China
| | - Zhe-Sheng Chen
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong PR China
| | - Zhenbo Hu
- Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261035, Shandong PR China
| | - Min Cui
- Department of Pain Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong PR China
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong PR China
| |
Collapse
|
33
|
Fatima S, Kumar V, Kumar D. Molecular mechanism of genetic, epigenetic, and metabolic alteration in lung cancer. Med Oncol 2025; 42:61. [PMID: 39893601 DOI: 10.1007/s12032-025-02608-5] [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] [Accepted: 01/13/2025] [Indexed: 02/04/2025]
Abstract
Lung cancer, a leading cause of cancer-related deaths worldwide, is primarily linked to smoking, tobacco use, air pollution, and exposure to hazardous chemicals. Genetic alterations, particularly in oncogenes like RAS, EGFR, MYC, BRAF, HER, and P13K, can lead to metabolic changes in cancer cells. These cells often rely on glycolysis for energy production, even in the presence of oxygen, a phenomenon known as aerobic glycolysis. This metabolic shift, along with other alterations, contributes to cancer cell growth and survival. To develop effective therapies, it's crucial to understand the genetic and metabolic changes that drive lung cancer. This review aims to identify specific genes associated with these metabolic alterations and screen phytochemicals for their potential to target these genes. By targeting both genetic and metabolic pathways, we hope to develop innovative therapeutic approaches to combat lung cancer.
Collapse
Affiliation(s)
- Sheeri Fatima
- School of Health Science and Technology (SoHST), UPES, Dehradun, Uttarakhand, 248007, India
| | - Vineet Kumar
- Chemistry & Bioprospecting Division, Forest Research Institute, Dehradun, 248006, India
| | - Dhruv Kumar
- School of Health Science and Technology (SoHST), UPES, Dehradun, Uttarakhand, 248007, India.
| |
Collapse
|
34
|
Kao WH, Chiu KY, Tsai SCS, Teng CLJ, Oner M, Lai CH, Hsieh JT, Lin CC, Wang HY, Chen MC, Lin H. PI3K/Akt inhibition promotes AR activity and prostate cancer cell proliferation through p35-CDK5 modulation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167568. [PMID: 39536992 DOI: 10.1016/j.bbadis.2024.167568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Aberrant PI3K/Akt activation is linked to prostate cancer (PCa) malignancy, while androgen receptor (AR) is critical in early-stage PCa development. Investigating the interaction between these pathways is crucial for PCa malignancy. Our previous study demonstrated that p35-CDK5 mediates post-translational modifications of AR, STAT3, and p21CIP1, eventually promoting PCa cell growth. This study revealed the role of p35-CDK5 in between PI3K/Akt and AR by utilizing LNCaP and 22Rv1 cells. Through the TCGA database analysis, we observed a positive correlation between PTEN and p35 expression, implying a potential negative correlation between PI3K/Akt activation and p35-CDK5. Inhibiting PI3K/Akt with LY294002, Capivasertib (AZD5363), or using an inactive Akt mutant significantly increased p35 expression and subsequently enhanced AR stability and activation in PCa cells. On the other hand, CDK5-knockdown reversed these effects. The involvement of the β-catenin/Egr1-axis was observed in regulating PI3K/Akt inhibition and p35-CDK5 activation, implying a possible mechanistic connection. Importantly, CDK5 knockdown further reduced PI3K/Akt-inhibition-induced AR and cell viability maintenance, suggesting a compensatory role for CDK5-AR in maintaining cell viability under Akt inhibition. In conclusion, PI3K/Akt inhibition could trigger p35-CDK5-dependent AR activation and cell viability, highlighting p35-CDK5 as a critical link connecting PI3K/Akt inhibition to AR activation and pivotal in PCa cell resistance to PI3K/Akt blockade.
Collapse
Affiliation(s)
- Wei-Hsiang Kao
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan; Translational Cell Therapy Center, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Kun-Yuan Chiu
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Stella Chin-Shaw Tsai
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan; Superintendent Office, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan; College of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | - Chieh-Lin Jerry Teng
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan; Division of Hematology/Medical Oncology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan.
| | - Muhammet Oner
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, TX75390, USA.
| | - Chi-Chien Lin
- Institute of Biomedical Science, National Chung Hsing University, Taichung 40227, Taiwan
| | - Hsin-Yi Wang
- Department of Nuclear Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan.
| | - Mei-Chih Chen
- Translational Cell Therapy Center, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
35
|
Jain A, Das R, Giri M, Mane P, Shard A. Carbohydrate kinase inhibition: a promising strategy in cancer treatment. Drug Discov Today 2025; 30:104308. [PMID: 39912130 DOI: 10.1016/j.drudis.2025.104308] [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: 12/09/2024] [Revised: 01/21/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025]
Abstract
Carbohydrate kinases (CKs) are pivotal in various biological processes, including energy consumption, cell signaling, and biosynthesis. They are a group of enzymes that facilitate the phosphorylation of carbohydrates, playing a crucial role in cellular metabolism. These enzymes facilitate the transfer of a phosphate group from a high-energy donor like ATP to a specified location on a carbohydrate substrate. Dysregulated kinase activity drives tumor growth and progression. Inhibitors targeting these enzymes have been developed and used in cancer therapy. The CK family encompasses three major types: hexokinases, ribokinases, and phosphatidylinositol kinases, with inhibitors of paramount importance in cancer treatment. This review explores the role of CKs in cancer and its inhibitors, providing insights into improving existing inhibitors and designing new ones.
Collapse
Affiliation(s)
- Archit Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Rudradip Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Muskan Giri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Pranita Mane
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
36
|
Gouda MA, Wei Z, Rodon J, Davies MA, Janku F, Gray RJ, Wang V, McShane LM, Rubinstein LV, Patton DR, Williams PM, Hamilton SR, Liu R, Bota DA, Swiecicki PL, Buchschacher GL, Tricoli JV, Conley BA, Arteaga CL, Harris LN, O’Dwyer PJ, Chen AP, Flaherty KT. Phase II Study of Copanlisib in Patients With PTEN Loss: Results From NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocols Z1G and Z1H. JCO Precis Oncol 2025; 9:e2400451. [PMID: 39913886 PMCID: PMC12002398 DOI: 10.1200/po-24-00451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/10/2024] [Accepted: 12/26/2024] [Indexed: 02/20/2025] Open
Abstract
PURPOSE Copanlisib, a pan-class phosphatidylinositol 3-kinase (PI3K) inhibitor with activity predominantly against the PI3K-delta and PI3K-alpha isoforms, has shown promising results in preclinical cancer models with PTEN loss. Herein, we report the activity and safety data from the Z1G and Z1H subprotocols, which included patients with PTEN loss, of the National Cancer Institute Molecular Analysis for Therapy Choice trial. METHODS Patients with complete loss of cytoplasmic and nuclear PTEN as determined by immunohistochemistry regardless of PTEN mutation or deletion status were included in subprotocol Z1G, and patients with a deleterious mutation in the PTEN gene and retained expression of PTEN were included in subprotocol Z1H. Copanlisib was given intravenously over 1 hour at a dose of 60 mg on days 1, 8, and 15 in a 21-day-on and 7-day-off schedule in 28-day cycles. Patients continued treatment until disease progression or unacceptable toxicity. RESULTS Overall, 49 patients (20 patients in Z1G and 29 in Z1H) were included in the primary efficacy analyses. The objective response rates in both cohorts were 0% (Z1G; 90% CI, 0 to 13.9) and 3.4% (Z1H; 90% CI, 0.2 to 15.3), respectively. The median progression-free and overall survival durations were 1.8 months (90% CI, 1.4 to 3.9 months) and 13.7 months (90% CI, 6.8 to 18.3 months) for the Z1G cohort and 1.8 months (90% CI, 1.8 to 2.1 months) and 9.0 months (90% CI, 5.4 to 13.3 months) for the Z1H cohort, respectively. CONCLUSION Our results do not support the antitumor activity of single-agent copanlisib in tumors with PTEN loss regardless of mutation or deletion status or PTEN deleterious mutations with PTEN expression.
Collapse
Affiliation(s)
| | - Zihan Wei
- Dana Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Jordi Rodon
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Filip Janku
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert J. Gray
- Dana Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Victoria Wang
- Dana Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Lisa M. McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Larry V. Rubinstein
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - David R. Patton
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Bethesda, MD
| | | | - Stanley R. Hamilton
- City of Hope National Medical Center and Comprehensive Cancer Center, Duarte, CA
| | - Raymond Liu
- Department of Hematology Oncology, The Permanente Medical Group, San Francisco, CA
| | - Daniela A. Bota
- UCI Health Chao Family Comprehensive Cancer Center, Orange, CA
| | | | | | - James V. Tricoli
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Barbara A. Conley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Lyndsay N. Harris
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Alice P. Chen
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | |
Collapse
|
37
|
Jia Y, Sun J, Chen S, Bian Y, Jiang A, Liang H, Du X. Dedicator of cytokinesis protein 2 activates the epithelial-mesenchymal transition in renal fibrosis through the Rac1/PI3K/AKT pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119894. [PMID: 39725220 DOI: 10.1016/j.bbamcr.2024.119894] [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: 09/01/2024] [Revised: 11/28/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Renal fibrosis is the most important feature of the progression of chronic kidney disease (CKD), and epithelial-mesenchymal transition (EMT) plays an important role in renal fibrosis. Dedicator of cytokinesis protein 2 (Dock2) is involved in the immune system and the development of a variety of fibrotic diseases. However, its specific role in renal fibrosis remains unclear. Therefore, in this study, we investigated the role and mechanism of Dock2 in renal fibrosis. We constructed an in vivo mouse model of unilateral ureteral obstruction (UUO) and an in vitro model of recombinant human transforming growth factor-β1 (TGF-β1)-induced HK-2 cells. The function and regulatory mechanism of Dock2 were studied via Western blotting, qRT-PCR, immunohistochemistry and immunofluorescence. First, Dock2 was more highly expressed in the kidneys of UUO mice than in those of sham-operated mice. A reduction in Dock2 can improve pathological changes in the kidney tissue of UUO mice, reduce the deposition of the extracellular matrix (ECM), and alleviate EMT. Silencing Dock2 reduced the activation of both the Rac1 pathway and the PI3K/AKT pathway. TGF-β1 promoted Dock2 expression in HK-2 cells in vitro. A decrease in Dock2 can inhibit the expression of Fibronectin, Collagen I, α-SMA and Vimentin and increase the level of E-cadherin. Treatment of HK-2 cells with the Rac1 activator 8-CPT or the PI3K/AKT pathway activator YS-49 inhibited the above changes induced by siDock2, indicating that Dock2 activates EMT in renal fibrosis through the Rac1/PI3K/AKT pathway. Our data suggest that Dock2 may be a potential target for renal fibrosis treatment.
Collapse
Affiliation(s)
- Yuanyuan Jia
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jing Sun
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Sha Chen
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yu Bian
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Anni Jiang
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Haihai Liang
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China; College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Xuanyi Du
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
38
|
Hashemi M, Mohandesi Khosroshahi E, Asadi S, Tanha M, Ghatei Mohseni F, Abdolmohammad Sagha R, Taheri E, Vazayefi P, Shekarriz H, Habibi F, Mortazi S, Khorrami R, Nabavi N, Rashidi M, Taheriazam A, Rahimzadeh P, Entezari M. Emerging roles of non-coding RNAs in modulating the PI3K/Akt pathway in cancer. Noncoding RNA Res 2025; 10:1-15. [PMID: 39296640 PMCID: PMC11406677 DOI: 10.1016/j.ncrna.2024.08.002] [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: 11/28/2023] [Revised: 07/25/2024] [Accepted: 08/08/2024] [Indexed: 09/21/2024] Open
Abstract
Cancer progression results from the dysregulation of molecular pathways, each with unique features that can either promote or inhibit tumor growth. The complexity of carcinogenesis makes it challenging for researchers to target all pathways in cancer therapy, emphasizing the importance of focusing on specific pathways for targeted treatment. One such pathway is the PI3K/Akt pathway, which is often overexpressed in cancer. As tumor cells progress, the expression of PI3K/Akt increases, further driving cancer advancement. This study aims to explore how ncRNAs regulate the expression of PI3K/Akt. NcRNAs are found in both the cytoplasm and nucleus, and their functions vary depending on their location. They can bind to the promoters of PI3K or Akt, either reducing or increasing their expression, thus influencing tumorigenesis. The ncRNA/PI3K/Akt axis plays a crucial role in determining cell proliferation, metastasis, epithelial-mesenchymal transition (EMT), and even chemoresistance and radioresistance in human cancers. Anti-tumor compounds can target ncRNAs to modulate the PI3K/Akt axis. Moreover, ncRNAs can regulate the PI3K/Akt pathway both directly and indirectly.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahsa Tanha
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Forough Ghatei Mohseni
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ramina Abdolmohammad Sagha
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elham Taheri
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Paria Vazayefi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Helya Shekarriz
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Habibi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shaghayegh Mortazi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Noushin Nabavi
- Independent Researchers, Victoria, British Columbia, V8V 1P7, Canada
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
39
|
Zhang Y, Zhou YL, Xu N, Meng T, Wang ZZ, Pan FM, Zhu LX. Chemokines and PI3K/AKT signaling pathway mediate the spontaneously ruptured hepatocellular carcinoma through the regulation of the cell cycle. Hepatobiliary Pancreat Dis Int 2025:S1499-3872(25)00029-3. [PMID: 39952875 DOI: 10.1016/j.hbpd.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/26/2024] [Indexed: 02/17/2025]
Abstract
BACKGROUND The incidence of spontaneously ruptured hepatocellular carcinoma (srHCC) has been shown to significantly elevate mortality rates. However, the precise mechanisms underlying srHCC remain poorly understood. METHODS Analysis was conducted on the data of 198 hepatocellular carcinoma (HCC) patients to investigate the factors contributing to srHCC. The clinical data of 33 transcriptome HCC patients were served for verification. An in-depth transcriptome analysis was conducted to investigate the distinctions between 26 cases of srHCC and 35 cases of non-ruptured hepatocellular carcinoma (nrHCC). Weighted Gene Co-expression Network Analysis (WGCNA) tool was utilized to develop a gene co-expression network. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathways enrichment, and protein-protein interaction (PPI) network were carried out. The corresponding samples for spontaneously ruptured hepatocellular carcinoma tissue (srHCC-T) and ruptured hepatocellular carcinoma paracancerous tissue (srHCC-P) was selected for verification. Transcriptional data were validated through reverse transcription quantitative polymerase chain reaction (RT-qPCR). Immunofluorescence (IF), immunohistochemistry (IHC) and Western blot were used to detect the protein expression. RESULTS Our results showed that white blood cell (WBC) and monocyte levels were significant independent risk factors for srHCC (P < 0.05). There was a strong association between the srHCC-T and the expression of cell cycle-related genes BUB1B and macrophage function-related gene MACRO. Furthermore, chemokines and the PI3K/AKT signaling pathway play a crucial role in regulating the cell cycle process through a complex network of interactions, ultimately impacting the occurrence of srHCC. CONCLUSIONS Our study confirms that chemokines and the PI3K/AKT signaling pathway mediate the occurrence of HCC rupture by regulating the cell cycle. We provide a theoretical basis for the clinical treatment of srHCC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of General Surgery, the Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Yang-Liu Zhou
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Na Xu
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Tao Meng
- Department of General Surgery, Hefei First People's Hospital, Hefei 230000, China
| | - Zhen-Zhen Wang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Fa-Ming Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei 230032, China.
| | - Li-Xin Zhu
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| |
Collapse
|
40
|
Lei J, Chen J, Yu W, Wu Q, Jing S, Tang Y, Lin L, Hu M. Portrait of WWP1: the current state in human cancer. Front Cell Dev Biol 2025; 12:1516613. [PMID: 39949609 PMCID: PMC11821962 DOI: 10.3389/fcell.2024.1516613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 12/31/2024] [Indexed: 02/16/2025] Open
Abstract
WWP1, a member of the C2-WW-HECT E3 ligase family, is an E3 ubiquitin-protein ligase containing WW domains. This enzyme plays a critical role in regulating diverse cellular processes. Its expression is modulated by various factors and non-coding RNAs, resulting in ubiquitination that affects substrate protein degradation. WWP1 demonstrates a dual function, acting predominantly as an oncogene in tumors but occasionally as a tumor suppressor. This review summarizes WWP1's biological roles, therapeutic potential in oncology, upstream regulatory factors, and downstream substrates. It aims to promote research on WWP1's antitumor effects, improve understanding of its role in tumorigenesis, and support the development of targeted therapies.
Collapse
Affiliation(s)
- Jiaming Lei
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Jun Chen
- The Central Hospital of Ezhou, Affiliated Hospital of Hubei University of Science and Technology, Ezhou, Hubei, China
| | - Wenwen Yu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Qing Wu
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shuang Jing
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yuanguang Tang
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Li Lin
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Meichun Hu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| |
Collapse
|
41
|
Mghwary AES, Hassan RA, Halim PA, Abdelhameid MK. Advances in structural identification of some thieno[2,3-d]pyrimidine scaffolds as antitumor molecules: Synthetic approaches and control programmed cancer cell death potential. Bioorg Chem 2025; 154:107985. [PMID: 39637483 DOI: 10.1016/j.bioorg.2024.107985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/30/2024] [Accepted: 11/17/2024] [Indexed: 12/07/2024]
Abstract
Thieno[2,3-d]pyrimidine fragment is not only bioistostere to quinazoline ring but also to purines which exist in nucleic acids responsible for several key biological processes of the living cells, thus it is of a great interest for many researchers. Thieno[2,3-d]pyrimidine ring has become an important scaffold for different compounds with versatile pharmacological activities including anticancer. These compounds exert their anticancer activity through variant mechanisms of action; one of these is the induction of different programmed cell death types as apoptosis and necroptosis which is an effective approach for cancer treatment. This review highlights the different synthetic approaches of recent thieno[2,3-d]pyrimidine analogs along with their anticancer significance through induction of apoptotic or necroptotic cell death with illustration of the structure-activity relationship (SAR).
Collapse
Affiliation(s)
- Aml E-S Mghwary
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Rasha A Hassan
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Peter A Halim
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Mohammed K Abdelhameid
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| |
Collapse
|
42
|
Yue L, Li N, Ye X, Xiu Y, Wang B. Polymethoxylated flavones for modulating signaling pathways in inflammation. Int Immunopharmacol 2024; 143:113522. [PMID: 39515044 DOI: 10.1016/j.intimp.2024.113522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/18/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Aberrant signaling pathways play a crucial role in the pathogenesis of various diseases, including inflammatory disorders and autoimmune conditions. Polymethoxylated flavones (PMFs), a class of natural compounds found in citrus fruits, have obtained increasing attention for their potential therapeutic effects in modulating inflammatory responses. Although significant progress has been made in the pharmacological research of PMFs, the mechanisms by which they modulate signaling pathways to treat inflammation have not been systematically reviewed or analyzed. To address this gap in the literature, this review explores the mechanisms underlying the anti-inflammatory properties of PMFs and their prospects as drugs for treating inflammatory diseases. We discuss the molecular targets and signaling pathways through which PMFs exert their anti-inflammatory effects, including NF-κB pathway, PI3K/Akt pathway, MAPK pathway, Nrf2 pathway, and regulation of inflammatory cytokine production. Furthermore, we highlight preclinical studies evaluating the efficacy of PMFs in various inflammatory conditions, such as rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and osteoarthritis (OA). Despite promising findings, challenges remain in optimizing the pharmacokinetic properties and therapeutic efficacy of PMFs for clinical use. Future research directions include elucidating the structure-activity relationships of PMFs, developing novel delivery strategies, and conducting large-scale clinical trials to validate their efficacy and safety profiles. Overall, PMFs represent a promising class of natural compounds with potential applications as anti-inflammatory drugs, offering novel therapeutic opportunities for managing inflammatory diseases.
Collapse
Affiliation(s)
- Lixia Yue
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Ning Li
- Shenzhen Research Institute, the Hong Kong University of Science and Technology, Shenzhen 518054, China
| | - Xianglu Ye
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yanfeng Xiu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Bing Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
43
|
Liu L, Graff SL, Wang Y. New Emerging Therapies Targeting PI3K/AKT/mTOR/PTEN Pathway in Hormonal Receptor-Positive and HER2-Negative Breast Cancer-Current State and Molecular Pathology Perspective. Cancers (Basel) 2024; 17:16. [PMID: 39796647 PMCID: PMC11718791 DOI: 10.3390/cancers17010016] [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: 11/08/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
In hormone receptor-positive and HER2-negative breast cancers, a growing number of revolutionary personalized therapies are in clinical use or trials, such as CDK4/6 inhibitors, immune checkpoint inhibitors, and PIK3CA inhibitors. Those treatment options are largely driven by the presence or absence of genomic alterations in the tumor. Therefore, molecular profiling is often performed during disease progression. The most encountered genomic alterations are in the PI3K/AKT/mTOR/PTEN pathway. This review discusses the genetic alterations associated with the PI3K/AKT/mTOR/PTEN pathway to help clinicians understand drug selection, resistance, or interaction from a molecular pathologist's perspective.
Collapse
Affiliation(s)
- Liu Liu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Brown University Health, Providence, RI 02903, USA;
- Legorreta Cancer Center, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| | - Stephanie L. Graff
- Legorreta Cancer Center, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
- Division of Medical Oncology, Rhode Island Hospital and Brown University Health, Providence, RI 02903, USA
| | - Yihong Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Brown University Health, Providence, RI 02903, USA;
- Legorreta Cancer Center, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| |
Collapse
|
44
|
Huete-Acevedo J, Mas-Bargues C, Arnal-Forné M, Atencia-Rabadán S, Sanz-Ros J, Borrás C. Role of Redox Homeostasis in the Communication Between Brain and Liver Through Extracellular Vesicles. Antioxidants (Basel) 2024; 13:1493. [PMID: 39765821 PMCID: PMC11672896 DOI: 10.3390/antiox13121493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Extracellular vesicles (EVs) are small, membrane-bound particles secreted by cells into the extracellular environment, playing an increasingly recognized role in inter-organ communication and the regulation of various physiological processes. Regarding the redox homeostasis context, EVs play a pivotal role in propagating and mitigating oxidative stress signals across different organs. Cells under oxidative stress release EVs containing signaling molecules that can influence the redox status of distant cells and tissues. EVs are starting to be recognized as contributors to brain-liver communication. Therefore, in this review, we show how redox imbalance can affect the release of EVs in the brain and liver. We propose EVs as mediators of redox homeostasis in the brain-liver axis.
Collapse
Affiliation(s)
- Javier Huete-Acevedo
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, 46010 Valencia, Spain; (J.H.-A.); (C.M.-B.); (M.A.-F.); (S.A.-R.)
| | - Cristina Mas-Bargues
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, 46010 Valencia, Spain; (J.H.-A.); (C.M.-B.); (M.A.-F.); (S.A.-R.)
| | - Marta Arnal-Forné
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, 46010 Valencia, Spain; (J.H.-A.); (C.M.-B.); (M.A.-F.); (S.A.-R.)
| | - Sandra Atencia-Rabadán
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, 46010 Valencia, Spain; (J.H.-A.); (C.M.-B.); (M.A.-F.); (S.A.-R.)
| | - Jorge Sanz-Ros
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Consuelo Borrás
- MiniAging Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, 46010 Valencia, Spain; (J.H.-A.); (C.M.-B.); (M.A.-F.); (S.A.-R.)
| |
Collapse
|
45
|
Xu L, Zhao X, Tang F, Zhang J, Peng C, Ao H. Ameliorative Effect of Ginsenoside Rc on 5-Fluorouracil-Induced Chemotherapeutic Intestinal Mucositis via the PI3K-AKT/NF-κB Signaling Pathway: In Vivo and In Vitro Evaluations. Int J Mol Sci 2024; 25:13085. [PMID: 39684797 DOI: 10.3390/ijms252313085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/26/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
5-Fluorouracil (5-Fu) is a chemotherapeutic agent widely used to treat various cancers, which causes intestinal mucositis as a common side effect. Ginsenoside Rc, an active compound with anti-inflammatory, antioxidant, immunomodulatory, and antitumor properties, has protective effects against chemotherapy-induced mucositis caused by 5-Fu. This study aims to evaluate the protective effects of Rc on 5-Fu-induced chemotherapy-related mucositis and to elucidate its underlying mechanisms. In vivo experiments were conducted to measure intestinal permeability and assess the effects of Rc on body weight loss, diarrhea, and intestinal pathology induced by 5-Fu. Network pharmacology was also employed to explore potential mechanisms. In vitro, IEC-6 cell models were used to validate the cytoprotective effects of Rc, including assessments of cell viability, apoptosis, lactate dehydrogenase (LDH) release, and changes in inflammatory cytokine levels. The results indicate that Rc significantly ameliorated body weight reduction, diarrhea, and intestinal damage in mice treated by 5-Fu. Rc significantly mitigated 5-Fu-induced cellular damage by reducing levels of inflammatory cytokines such as IL-1β, IL-6, and TNF-α and decreasing apoptosis and cell permeability. Western blot analysis revealed that Rc upregulated the expression of Bcl-2 and tight junction proteins and downregulated the expression of Bax. Furthermore, Rc exerts anti-inflammatory and anti-apoptotic effects through PI3K-AKT and NF-κB signaling pathways. In conclusion, ginsenoside Rc demonstrated significant protective effects against 5-Fu-induced intestinal mucositis via the PI3K-AKT/NF-κB signaling pathway, suggesting its potential as a therapeutic agent for chemotherapy-related mucositis.
Collapse
Affiliation(s)
- Liyue Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaolan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jingnan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu, University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
46
|
Khalafiyan A, Fadaie M, Khara F, Zarrabi A, Moghadam F, Khanahmad H, Cordani M, Boshtam M. Highlighting roles of autophagy in human diseases: a perspective from single-cell RNA sequencing analyses. Drug Discov Today 2024; 29:104224. [PMID: 39521332 DOI: 10.1016/j.drudis.2024.104224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Autophagy, the lysosome-driven breakdown of intracellular components, is pivotal in regulating eukaryotic cellular processes and maintaining homeostasis, making it physiologically important even under normal conditions. Cellular mechanisms involving autophagy include the response to nutrient deprivation, intracellular quality control, early development, and cell differentiation. Despite its established health significance, the role of autophagy in cancer and other diseases remains complex and not fully understood. A comprehensive understanding of autophagy is crucial to facilitate the development of novel therapies and drugs that can protect and improve human health. High-throughput technologies, such as single-cell RNA sequencing (scRNA-seq), have enabled researchers to study transcriptional landscapes at single-cell resolution, significantly advancing our knowledge of autophagy pathways across diverse physiological and pathological contexts. This review discusses the latest advances in scRNA-seq for autophagy research and highlights its potential in the molecular characterization of various diseases.
Collapse
Affiliation(s)
- Anis Khalafiyan
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahmood Fadaie
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Khara
- Department of Biology, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Fariborz Moghadam
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
47
|
Zhu S, Yu D, Wang X, Wang X. Predict the Drug-Drug Interaction of a Novel PI3Kα/δ Inhibitor, TQ-B3525, and Its Two Metabolites Using Physiologically Based Pharmacokinetic Modeling. J Clin Pharmacol 2024; 64:1517-1527. [PMID: 39105511 DOI: 10.1002/jcph.6111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024]
Abstract
A novel dual PI3K α/δ inhibitor, TQ-B3525, has been developed for the targeted treatment of lymphoma and solid tumors. TQ-B3525 is primarily metabolized by CYP3A4 and FOM3, while also serving as a substrate for the P-glycoprotein transporter. The aim of this study was to anticipate the drug-drug interaction (DDI) of TQ-B3525 and its two metabolites with CYP3A4 enzyme potent inducer (rifampicin) and CYP3A4/P-gp inhibitor (itraconazole) utilizing a physiologically based pharmacokinetic (PBPK) modeling approach. Clinical data from healthy and cancer patient adults were employed to construct and evaluate the PBPK model for TQ-B3525, M3, and M8-3. Models involving rifampicin combined with midazolam, itraconazole combined with midazolam or digoxin were utilized to showcase the robustness of evaluating DDI effects. The simulated drug exposure of TQ-B3525, M3, and M8-3 in healthy and patient adults were consistent with clinical data, and the mean fold error values were within the acceptable ranges. The simulated results of positive substrates correspond to those reported in the literature. Co-administration with rifampicin reduces Cmax and AUC of TQ-B3525 to 76.1% and 46.0%, while increasing the levels of M3 and M8-3. With itraconazole, Cmax and AUC of TQ-B3525 rise to 131% and 204%, but decrease substantially for M3 and M8-3. PBPK model simulation results showed that the systemic exposure of TQ-B3525 was significantly affected when co-administered with CYP3A4/P-gp inducers and inhibitors. This indicates that the combination with strong inducers and inhibitors should be carefully avoided or adjust the dosage of TQ-B3525 in clinic.
Collapse
Affiliation(s)
- Shixing Zhu
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| | - Ding Yu
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| | - Xunqiang Wang
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| | - Xin Wang
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| |
Collapse
|
48
|
Yuan HY, Liu WY, Feng G, Chen SD, Jin XZ, Chen LL, Song ZJ, Li K, Byrne CD, Targher G, Tian N, Li G, Zhang XL, George J, Zhou M, Wang F, Zheng MH. Associations between cuprotosis-related genes and the spectrum of metabolic dysfunction-associated fatty liver disease: An exploratory study. Diabetes Obes Metab 2024; 26:5757-5775. [PMID: 39285685 DOI: 10.1111/dom.15946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 11/05/2024]
Abstract
AIMS To explore the associations between cuprotosis-related genes (CRGs) across different stages of liver disease in metabolic dysfunction-associated fatty liver disease (MAFLD), including hepatocellular carcinoma (HCC). MATERIALS AND METHODS We analysed several bulk RNA sequencing datasets from patients with MAFLD (n = 331) and MAFLD-related HCC (n = 271) and two MAFLD single-cell RNA sequencing datasets. To investigate the associations between CRGs and MAFLD, we performed differential correlation, logistic regression and functional enrichment analyses. We also validated the findings in an independent Wenzhou PERSONS cohort of MAFLD patients (n = 656) used for a genome-wide association study (GWAS). RESULTS GLS, GCSH and ATP7B genes showed significant differences across the MAFLD spectrum and were significantly associated with liver fibrosis stages. GLS was closely associated with fibrosis stages in patients with MAFLD and those with MAFLD-related HCC. GLS is predominantly expressed in monocytes and T cells in MAFLD. During the progression of metabolic dysfunction-associated fatty liver to metabolic-associated steatohepatitis, GLS expression in T cells decreased. GWAS revealed that multiple single nucleotide polymorphisms in GLS were associated with clinical indicators of MAFLD. CONCLUSIONS GLS may contribute to liver inflammation and fibrosis in MAFLD mainly through cuprotosis and T-cell activation, promoting the progression of MAFLD to HCC. These findings suggest that cuprotosis may play a role in MAFLD progression, potentially providing new insights into MAFLD pathogenesis.
Collapse
Affiliation(s)
- Hai-Yang Yuan
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wen-Yue Liu
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gong Feng
- Xi'an Medical University, Xi'an, China
| | - Sui-Dan Chen
- Department of Pathology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin-Zhe Jin
- Department of Laboratory Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li-Li Chen
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zi-Jun Song
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Li
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Christopher D Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton General Hospital, Southampton, UK
| | - Giovanni Targher
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Na Tian
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gang Li
- Department of Infectious, Jining No.1 People's Hospital, Jining, China
| | - Xin-Lei Zhang
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jacob George
- Storr Liver Centre, Westmead Hospital and University of Sydney, Westmead, Australia
| | - Meng Zhou
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| |
Collapse
|
49
|
Wang Z, Guo Y, Li K, Huo Y, Wang S, Dong S, Ma M. Targeting the PI3K/mTOR pathway in idiopathic pulmonary fibrosis: Advances and therapeutic potential. Bioorg Med Chem 2024; 115:117908. [PMID: 39471771 DOI: 10.1016/j.bmc.2024.117908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 11/01/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, fatal lung disease characterized by irreversible tissue scarring, leading to severe respiratory dysfunction. Despite current treatments with the drugs Pirfenidone and Nintedanib, effective management of IPF remains inadequate due to limited therapeutic benefits and significant side effects. This review focuses on the phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway, a critical regulator of cellular processes linked to fibrosis, such as fibroblast proliferation, inflammation, and epithelial-mesenchymal transition (EMT). We discuss recent advances in understanding the role of the PI3K/mTOR pathway in IPF pathogenesis and highlight emerging therapies targeting this pathway. The review compiles evidence from both preclinical and clinical studies, suggesting that PI3K/mTOR inhibitors may offer new hope for IPF treatment by modulating fibrosis and improving patient outcomes. Moreover, it outlines the potential for these inhibitors to be developed into effective, personalized treatment options, underscoring the importance of further research to explore their efficacy and safety profiles comprehensively.
Collapse
Affiliation(s)
- Zhengyang Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Yanzhi Guo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Kaiyin Li
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Yan Huo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Shuyan Wang
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Suzhen Dong
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China.
| | - Mingliang Ma
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China; Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
50
|
Liu M, Liu Q, Hu K, Dong Y, Sun X, Zou Z, Ji D, Liu T, Yu Y. Colorectal cancer with BRAF V600E mutation: Trends in immune checkpoint inhibitor treatment. Crit Rev Oncol Hematol 2024; 204:104497. [PMID: 39245296 DOI: 10.1016/j.critrevonc.2024.104497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/10/2024] Open
Abstract
Colorectal cancer (CRC) with BRAF V600E mutation presents a formidable scientific and clinical challenge due to its aggressive nature and poor response to standard therapeutic approaches. BRAF V600E mutation-induced conspicuous activation of the MAPK pathway contributes to the relentless tumor progression. Nevertheless, the efficacy of multi-targeted MAPK pathway inhibition remains suboptimal in clinical practice. Patients with high microsatellite instability (MSI-H) have shown favorable results with immune checkpoint inhibitors (ICIs). The combination of the MAPK pathway inhibition with ICIs has recently emerged as a promising regimen to improve clinical outcomes in the microsatellite stable (MSS) subgroup of BRAF V600E-mutant metastatic CRC patients. In this review, we elucidate the unique tumor biology of BRAF V600E-mutant CRC, with a particular focus on the immune features underlying the rationale for ICI treatments in the MSI-H and MSS subpopulations, then highlight the trends in clinical trials of the ICI therapy for BRAF V600E-mutant metastatic CRC.
Collapse
Affiliation(s)
- Mengling Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qing Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Keshu Hu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yu Dong
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xun Sun
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhiguo Zou
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Dingkun Ji
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|