1
|
Xu NN, Wen YC, Pan J, Shu F, Qu JX, Qi XF, Tang J. Activated mTORC1 signaling pathway aggravates cisplatin induced oxidative damage by inhibiting autophagy in mouse cochlear hair cells. Neuropharmacology 2025; 272:110433. [PMID: 40147638 DOI: 10.1016/j.neuropharm.2025.110433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Platinum-based antitumor drugs, such as cisplatin and carboplatin, are well-known for their severe ototoxicity. The ototoxic effects of these drugs are primarily attributed to oxidative stress induced damage within cochlear hair cells (HCs), leading to cell death and subsequent irreversible hearing loss. Over the past decade, studies have demonstrated that upregulating autophagy levels in HCs can greatly alleviate the death of cochlear HCs as part of the oxidative damage induced by ototoxic drugs. However, the molecular mechanisms by which platinum-based drugs affect autophagy and ultimately lead to HCs death remain unclear. In the present study, we investigated the effects of cisplatin on the mTOR signaling pathway, a critical regulator of autophagy, in cochlear explants of mice. Our results indicated that while cisplatin enhances autophagy activity initially, it also activates mTOR Complex1 (mTORC1) within HCs. The persistent activation of mTORC1 inhibits autophagy in HCs, resulting in the accumulation of reactive oxygen species and leading to cell death. Further pharmacological experiments confirmed the protective role of rapamycin, a specific mTORC1 inhibitor, highlighting the importance of autophagy in combating cisplatin-induced ototoxicity. Our findings suggest that modulating the mTOR signaling pathway to regulate autophagy could be an effective strategy for preventing cisplatin-induced ototoxic damage.
Collapse
Affiliation(s)
- Na-Na Xu
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yin-Chuan Wen
- Department of Anesthesiology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518000, China
| | - Jing Pan
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Fan Shu
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jia-Xi Qu
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xiao-Fei Qi
- Department of Anesthesiology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518000, China.
| | - Jie Tang
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, China; Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Qiao Z, Feng X, Sun W, Wang F, Lu C. Independent and synergistic effects of extreme heat and NO 2 pollution on diabetic nephropathy in a type II diabetes mouse model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 375:126321. [PMID: 40294690 DOI: 10.1016/j.envpol.2025.126321] [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: 11/10/2024] [Revised: 04/13/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025]
Abstract
Extreme heat and traffic-related air pollution (TRAP) have been linked to worsening chronic health disorders, however, their combined effects on diabetic nephropathy (DN) are little understood. Type II diabetic mice were exposed to heat (40 °C) and NO2 (5 ppm) separately for 4 h per day over 6 weeks to investigate the synergistic effects on the progression of DN. We found that exposure to high temperature and NO2 elevated blood glucose levels and exacerbated histopathological changes. Additionally, there were increased oxidation indicators (ROS, MDA, 8-OHdG) and decreased antioxidant indicators (CAT, SOD, GSH-PX), along with elevated inflammation markers (TNF-α, IL-1β, IL-6). The expressions of transient receptor potential (TRP) ion channels (TRPV1, TRPV4, TRPA1, TRPM2) were also upregulated. Our findings suggest that simultaneous exposure to high temperature and NO2 impairs metabolic and autophagy pathways. Exposure to both high temperature and NO2 produces a synergistic effect, leading to more severe damage than exposure to either factor individually. This resulted in increased expression of APOA1, P62, and p-mTOR/mTOR while decreasing the expression of p-AMPKα/AMPKα and LC3-II/I. This disruption promoted the progression of DN. In contrast, capsazepine (CZP) reduced TRP expression, inflammatory markers, oxidative stress, metabolic and autophagy disorders, thereby mitigating renal damage and alleviating the progression of diabetic nephropathy. Our study provides some potential strategies for early prevention and effective reduction of DN.
Collapse
Affiliation(s)
- Zipeng Qiao
- XiangYa School of Public Health, Central South University, Changsha, 410013, China
| | - Xiangling Feng
- XiangYa School of Public Health, Central South University, Changsha, 410013, China
| | - Wenying Sun
- XiangYa School of Public Health, Central South University, Changsha, 410013, China
| | - Faming Wang
- Centre for Molecular Biosciences and Non-communicable Diseases, Xi'an University of Science and Technology, Xi'an, 710054, China
| | - Chan Lu
- XiangYa School of Public Health, Central South University, Changsha, 410013, China; FuRong Laboratory, Changsha, 410078, Hunan, China; Hunan Provincial Key Laboratory of Low Carbon Healthy Building, Central South University, Changsha, 410083, China.
| |
Collapse
|
3
|
Xu Y, Gu X, Li W, Lin B, Xu Y, Wei Q, Liu Q, Zhao Y, Long R, Jiang H, Wu Z, Liu Y, Qiang L. Autophagic degradation of SQSTM1 enables fibroblast activation to accelerate wound healing. Autophagy 2025. [PMID: 40400126 DOI: 10.1080/15548627.2025.2508546] [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: 09/03/2024] [Revised: 05/07/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025] Open
Abstract
Wound healing is a meticulously coordinated and intricate progression that necessitates precise regulation of fibroblast behavior. Macroautophagy/autophagy is a degradation system for clearing damaged cellular components. SQSTM1/p62 (sequestosome 1), a well-established autophagy receptor, also functions as a signaling hub beyond autophagy. Here, we observed a significant upregulation of autophagy in fibroblasts after wounding. Using mice with fibroblast-specific deletion of Atg7 (autophagy related 7), we found that fibroblast autophagy governed wound healing. Fibroblast autophagy deficiency delayed proper dermal repair that was mired in insufficient fibroblast proliferation, migration, and myofibroblast transition. In vitro experiments further revealed that autophagy deficiency disrupted TGFB1 (transforming growth factor beta 1)-induced fibroblast proliferation, migration, and myofibroblast differentiation. Mechanistically, autophagy deficiency led to SMAD2 (SMAD family member 2) and SMAD3 sequestration within SQSTM1 bodies and attenuated TGFB1-induced receptor-regulated SMAD (R-SMAD) phosphorylation in an SQSTM1-dependent manner. Furthermore, sqstm1 deletion rescued the delayed skin wound healing caused by autophagy deficiency, and autophagy inducers promoted wound healing in an SQSTM1-dependent manner. Our findings highlight the critical role of fibroblast autophagy in wound healing and elucidate the underlying mechanisms by which autophagy regulates fibroblast behavior.
Collapse
Affiliation(s)
- Yujiao Xu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin Gu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenshu Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Boyang Lin
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yiting Xu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qingcheng Wei
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qingyuan Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yamin Zhao
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rongzhuo Long
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hulin Jiang
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Zhaoqiu Wu
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yunyao Liu
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lei Qiang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
4
|
Li X, Han Z, Ai J. Synergistic targeting strategies for prostate cancer. Nat Rev Urol 2025:10.1038/s41585-025-01042-6. [PMID: 40394240 DOI: 10.1038/s41585-025-01042-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2025] [Indexed: 05/22/2025]
Abstract
Prostate cancer is the second most commonly diagnosed cancer and the fifth leading cause of death among men worldwide. Androgen deprivation therapy is a common prostate cancer treatment, but its efficacy is often hindered by the development of resistance, which results in reducing survival benefits. Immunotherapy showed great promise in treating solid tumours; however, clinically significant improvements have not been demonstrated for patients with prostate cancer, highlighting specific drawbacks of this therapeutic modality. Hence, exploring novel strategies to synergistically enhance the efficacy of prostate cancer immunotherapy is imperative. Clinical investigations have focused on the combined use of targeted or gene therapy and immunotherapy for prostate cancer. Notably, tumour-specific antigens and inflammatory mediators are released from tumour cells after targeted or gene therapy, and the recruitment and infiltration of immune cells, including CD8+ T cells and natural killer cells activated by immunotherapy, are further augmented, markedly improving the efficacy and prognosis of prostate cancer. Thus, immunotherapy, targeted therapy and gene therapy could have reciprocal synergistic effects in prostate cancer in combination, resulting in a proposed synergistic model encompassing these three therapeutic modalities, presenting novel potential treatment strategies for prostate cancer.
Collapse
Affiliation(s)
- Xuanji Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Zeyu Han
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Chen T, Xu Y, Yang F, Pan Y, Ji N, Li J, Zeng X, Chen Q, Jiang L, Shen YQ. Crosstalk of glutamine metabolism between cancer-associated fibroblasts and cancer cells. Cell Signal 2025; 133:111874. [PMID: 40381975 DOI: 10.1016/j.cellsig.2025.111874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/06/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Glutamine (Gln), a critical metabolic substrate, fuels the uncontrolled proliferation of cancer cells. Cancer-associated fibroblasts (CAFs), essential components of the tumor microenvironment, facilitate tumor progression by supplying Gln to cancer cells and driving drug resistance through metabolic reprogramming. This review highlights the key processes of Gln uptake, transport, and catabolism and explores the metabolic crosstalk between CAFs and cancer cells. It also examines the roles of major oncogenic regulators-c-Myc, mTORC, KRAS, p53, and HIF-in controlling Gln metabolism and shaping therapeutic resistance. Current pharmacological approaches targeting Gln metabolism, including enzyme inhibitors and transporter blockers, are discussed alongside emerging therapeutic strategies and ongoing clinical trials. Lastly, we underscore the importance of integrating advanced technologies like artificial intelligence and spatial omics to refine treatment targeting and develop more effective, personalized therapeutic interventions.
Collapse
Affiliation(s)
- Tingyu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yiming Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fan Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanxin Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lu Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Mancini C, Menegazzi G, Peppicelli S, Versienti G, Guasti D, Pieraccini G, Rovida E, Lulli M, Papucci L, Dello Sbarba P, Biagioni A. BCR::ABL1 expression in chronic myeloid leukemia cells in low oxygen is regulated by glutamine via CD36-mediated fatty acid uptake. Cancer Cell Int 2025; 25:176. [PMID: 40369538 PMCID: PMC12080266 DOI: 10.1186/s12935-025-03805-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 05/04/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Chronic myeloid leukemia (CML) is influenced by microenvironmental nutrients, glucose (Glc), and glutamine (Gln) which regulate cell proliferation, viability, and the expression of the driver oncoprotein (BCR::ABL1). RESULTS Our study revealed that Glc, while partially supporting alone cell growth in normoxia, is essential in low oxygen conditions, whereas Gln is ineffective. Under low oxygen, Gln reduced oxidative respiratory activity while enhancing glycolysis. In these conditions, fatty acid (FA) metabolism becomes crucial, as evidenced by increased lipid droplets (LD) accumulation when Glc was absent. Gln, in particular, drives CD36-mediated FA uptake, suppressing the BCR::ABL1 oncoprotein and facilitating cell survival. By co-culturing leukemia cells with adipocytes, one of the main bone marrow (BM) cell components, we observed an enhanced FA release, suggesting a link between FA, microenvironmental BM cells, and the maintenance of leukemic stem cells (LSC). METHODS K562 and KCL22 cell lines were subjected to Glc and/or Gln deprivation under hypoxic conditions (96 h at 0.1% O2). Metabolic profiling was conducted through the Seahorse XFe96 analyzer, and the contribution of L-Glutamine-13C5 to FA de novo synthesis was determined via GC/MS. Intracellular neutral LD were measured using BODIPY 493/503 in confocal microscopy and flow cytometry, with their presence and morphology further examined via transmission electron microscopy. BCR::ABL1 as well as several FA-related markers were evaluated via Western Blotting, whilst CD36 was determined through flow cytometry. LC2 assay was used for measuring leukemia stem cell potential by inhibiting FA uptake via the usage of the Sulfo-N-Succinimidyl Oleate, a CD36 inhibitor. qPCR was exploited to detect markers of FA secretion in CML-adipocytes co-culture together with Nile Red staining to assess free FA in the media. CONCLUSIONS These findings underscore the central role of FA in the regulation of the LSC compartment of CML, highlighting the importance of Gln in facilitating CML cell survival under restrictive metabolic conditions and preparing the cell population for expansion upon the release of these restrictions.
Collapse
Affiliation(s)
- Caterina Mancini
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Giulio Menegazzi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Giampaolo Versienti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Giuseppe Pieraccini
- CISM Mass Spectrometry Center, Università degli Studi di Firenze, Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy.
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy.
| |
Collapse
|
7
|
Senapati PK, Mahapatra KK, Singh A, Bhutia SK. mTOR inhibitors in targeting autophagy and autophagy-associated signaling for cancer cell death and therapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189342. [PMID: 40339669 DOI: 10.1016/j.bbcan.2025.189342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
The mechanistic target of rapamycin (mTOR) is a protein kinase that plays a central regulatory switch to control multifaceted cellular processes, including autophagy. As a nutrient sensor, mTOR inhibits autophagy by phosphorylating and inactivating key regulators, including ULK1, Beclin-1, UVRAG, and TFEB, preventing autophagy initiation and lysosomal biogenesis. It also suppresses autophagy-related protein expression, prioritizing growth over cellular recycling. Under nutrient deprivation, mTORC1 activity decreases, allowing autophagy to restore cellular homeostasis. Hyperautophagic activities lead to autophagic cell death; sometime after the point of no return, the cell goes for non-apoptotic, non-necrotic cell death i.e., Autosis. In cancer, the crosstalk between autophagy and mTOR is context-dependent, driving either cell survival or autophagy-dependent cell death. Using mTOR inhibitors, autophagic cell death can be induced to regulate cell growth, and proliferation is a potential therapeutic option for cancer treatment. mTOR inhibitors are broadly categorized into two types, i.e., natural and synthetic mTOR inhibitors. Although several studies in preclinical and clinical trials of various synthetic mTOR inhibitors are now in focus for cancer therapies, limited work has been done to explore autophagic cell death-inducing mTOR inhibitors. In addition, many natural mTOR inhibitors display better efficacy over synthetic mTOR inhibitors due to their lower toxicity, biocompatibility, and potential to overcome drug resistance in inducing autophagic cell death for cancer treatment.
Collapse
Affiliation(s)
- Prakash Kumar Senapati
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Kewal Kumar Mahapatra
- Department of Agriculture and Allied Sciences (Zoology), C. V. Raman Global University Bhubaneswar, Odisha-752054, India
| | - Amruta Singh
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India.
| |
Collapse
|
8
|
Ou H, Sun B, Jiang Y, Gao M. Tanshinone IIA ameliorates septic immunosuppression by balancing the Th17/Treg cell ratio and modulating Treg function after induction of autophagy. Arch Biochem Biophys 2025; 770:110451. [PMID: 40334959 DOI: 10.1016/j.abb.2025.110451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/28/2025] [Accepted: 05/04/2025] [Indexed: 05/09/2025]
Abstract
Sepsis is a systemic inflammatory response syndrome due to infection with a high incidence and high mortality. The Tanshinone IIA (TSN) is an active ingredient extracted from the traditional Chinese medicine Danshen. This work attempted to investigate the functional role of TSN is sepsis. It was found that the ratio of Th17/Treg was increased in sepsis patients and mouse model. TSN reduced the ratio of Th17/Treg and the levels of IL-17, IL-23, TGF-β1, and IL-10 in CD4+ T cells. Proliferation of CD4+CD25-Teff cells was inhibited by TSN. The expression of co-stimulatory molecules ICOS and CTLA-4 was decreased in CD4+ cells following TSN treatment. Additionally, TSN elevated LC3-II/I expression and down-regulated p62, p-mTOR and p-p70S6K to activate autophagy. Autophagy activator rapamycin (Rapa) activated autophagy to reduce the ratio of Th17/Treg in CD4+ T cells. Autophagy inhibitor (3-MA) in activated autophagy to elevate the ratio of Th17/Treg in CD4+ T cells, which was abolished by TSN treatment. TSN alleviated the damage of kidney, lung and liver tissues and inhibited the ratio of Th17/Treg in sepsis mice by activating autophagy. In summary, this work demonstrated that TSN ameliorates sepsis-associated immunosuppression by activating autophagy and balancing the Th17/Treg cell ratio.
Collapse
Affiliation(s)
- Hao Ou
- Department of Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Bei Sun
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Jiang
- Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Institute of Emergency Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Min Gao
- Department of Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Zhang W, Jia X, Lou D, Wu Q, Yan Y, Mao F. Comparison of mTOR inhibitors combined with endocrine therapy versus that alone in breast cancer: a meta-analysis. Future Oncol 2025; 21:1417-1427. [PMID: 40152674 PMCID: PMC12051556 DOI: 10.1080/14796694.2025.2485022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND This meta-analysis aims to evaluate the efficacy and safety of rapamycin (mTOR) inhibitors with endocrine therapy versus endocrine therapy alone in treating advanced or metastatic estrogen receptor/progesterone receptor (ER/PR) + breast cancer. METHODS We conducted a comprehensive search in PubMed, Web of Science, Embase, and the Cochrane Library for randomized controlled trials (RCTs) comparing mTOR inhibitors plus endocrine therapy with endocrine therapy alone up to September 2024. RESULTS This analysis included 10 RCTs comprising 3,337 patients. Relative to endocrine therapy alone, the combination of mTOR inhibitors and endocrine therapy significantly improved the clinical benefit rate (RR = 1.41, p < 0.001), overall response rate (RR = 1.40, p = 0.006), progression-free survival (PFS; HR = 0.67, p < 0.001), and overall survival (OS; HR = 0.86, p = 0.056), although the improvement in OS was not statistically significant. Subgroup analyses indicated a more pronounced PFS advantage in patients under 65 years of age (HR = 0.55, p = 0.013) and those who had previously received chemotherapy (HR = 0.51, p = 0.001). However, the incidence of adverse events was higher in the combination therapy group, notably stomatitis (p < 0.001), elevated aspartate aminotransferase/alanine aminotransferase (p = 0.04), and diarrhea (p = 0.01). CONCLUSIONS The combination of mTOR inhibitors with endocrine therapy offers superior efficacy with manageable toxicities in patients with advanced or metastatic ER/PR+ breast cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Xinru Jia
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Dandi Lou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Qingping Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yici Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Feiyan Mao
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
10
|
Yan Z, He L, Yuan J, Niu Y, Shuai S, Luo S, Du C, Rao H. The splicing factor SRRM2 modulates two S6K kinases to promote colorectal cancer growth. Oncogene 2025; 44:1284-1299. [PMID: 39956864 DOI: 10.1038/s41388-025-03307-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
The mechanistic target of rapamycin (mTOR) pathway plays a critical role in cell growth and metabolic homeostasis. The ribosomal protein S6 kinases S6K1 and S6K2 are the major effectors of the mTOR pathway key to translation efficiency, but the underlying regulatory mechanisms remain largely unclear. In this study, we searched for mTOR regulators and found that the splicing factor SRRM2 modulates the levels of S6K1 and S6K2, thereby activating the mTOR-S6K pathway. Interestingly, SRRM2 facilitates the expression of S6K2 by modulating alternative splicing, and enhances the stability of the S6K1 protein by regulating the E3 ubiquitin ligase WWP2. Moreover, SRRM2 is highly expressed in colorectal cancer (CRC) tissues and is associated with a poor prognosis. SRRM2 promotes CRC growth in vitro and in vivo. Combined, these data reveal an oncogenic role of SRRM2 in CRC through activating the mTOR-S6K pathway by two different approaches, further suggesting SRRM2 as a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Zhengwei Yan
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Luling He
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jiawei Yuan
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yulong Niu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Shimin Shuai
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shiwen Luo
- School of Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Changzheng Du
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Beijing Tsinghua Changgung Hospital & Tsinghua University School of Medicine, 168 Litang Road, Changping District, Beijing, 102218, PR China
| | - Hai Rao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
11
|
Boff MO, Xavier FAC, Diz FM, Gonçalves JB, Ferreira LM, Zambeli J, Pazzin DB, Previato TTR, Erwig HS, Gonçalves JIB, Bruzzo FTK, Marinowic D, da Costa JC, Zanirati G. mTORopathies in Epilepsy and Neurodevelopmental Disorders: The Future of Therapeutics and the Role of Gene Editing. Cells 2025; 14:662. [PMID: 40358185 PMCID: PMC12071303 DOI: 10.3390/cells14090662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 05/15/2025] Open
Abstract
mTORopathies represent a group of neurodevelopmental disorders linked to dysregulated mTOR signaling, resulting in conditions such as tuberous sclerosis complex, focal cortical dysplasia, hemimegalencephaly, and Smith-Kingsmore Syndrome. These disorders often manifest with epilepsy, cognitive impairments, and, in some cases, structural brain anomalies. The mTOR pathway, a central regulator of cell growth and metabolism, plays a crucial role in brain development, where its hyperactivation leads to abnormal neuroplasticity, tumor formation, and heightened neuronal excitability. Current treatments primarily rely on mTOR inhibitors, such as rapamycin, which reduce seizure frequency and tumor size but fail to address underlying genetic causes. Advances in gene editing, particularly via CRISPR/Cas9, offer promising avenues for precision therapies targeting the genetic mutations driving mTORopathies. New delivery systems, including viral and non-viral vectors, aim to enhance the specificity and efficacy of these therapies, potentially transforming the management of these disorders. While gene editing holds curative potential, challenges remain concerning delivery, long-term safety, and ethical considerations. Continued research into mTOR mechanisms and innovative gene therapies may pave the way for transformative, personalized treatments for patients affected by these complex neurodevelopmental conditions.
Collapse
Affiliation(s)
- Marina Ottmann Boff
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Fernando Antônio Costa Xavier
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Medicine and Health Sciences, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Fernando Mendonça Diz
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Júlia Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Laura Meireles Ferreira
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Jean Zambeli
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, University of the Valley of the Rio dos Sinos (UNISINOS), São Leopoldo 93022-750, RS, Brazil
| | - Douglas Bottega Pazzin
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Thales Thor Ramos Previato
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Helena Scartassini Erwig
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Health and Life, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Fernanda Thays Konat Bruzzo
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Daniel Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Health and Life, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Jaderson Costa da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Gabriele Zanirati
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| |
Collapse
|
12
|
Ei ZZ, Racha S, Chunhacha P, Yokoya M, Moriue S, Zou H, Chanvorachote P. Substituents introduction of methyl and methoxy functional groups on resveratrol stabilizes mTOR binding for autophagic cell death induction. Sci Rep 2025; 15:14675. [PMID: 40287470 PMCID: PMC12033263 DOI: 10.1038/s41598-025-98616-6] [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: 12/15/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
The regulation of the mammalian target of rapamycin (mTOR) protein by cancer cells can lead to uncontrol of cancer cell growth and cancer therapy resistance. The drug discovery of the anticancer agent 5-(3-hydroxy-4-methoxyphenethyl)-2-methoxy-3-methylphenol (SM-3), a derivative of resveratrol by substituting a methyl group at the hydroxy group of ring A and adding a methoxy group at the para position of ring B, shows promising potential for targeting autophagy to induce cell death and suppress cancer stem cells (CSCs) through the inhibition of the mTOR protein. In human lung cancer cells, SM-3 showed greater efficacy, with lower IC50 values of 72.74 ± 0.13, 67.66 ± 0.10, and 43.24 ± 0.11 µM in A549, H292, and H460 cells, respectively, compared to the parent compound, Resveratrol (Res). Moreover, the selectivity index (SI) values for BEAS2B cells compared to tumor cells treated with SM-3 were 10.99, 11.81, and 18.49 for A549, H292, and H460 cell lines, respectively. Therefore, SM-3 treatment led to reduced proliferation rates and colony formation in lung cancer cells. In our study, spheroids treated with SM-3 showed a higher proportion of dead spheroids compared to those treated with Res. Additionally, SM-3 treatment resulted in decreased expression of stem cell markers (CD133, CD44, and ALDH1A1) and transcription factors (OCT4, NANOG, and SOX2) in spheroids and organoids from human lung cancer cells by inhibiting the mTOR/pAkt pathway. SM-3 was also found to induce autophagic cell death, as indicated by Monodansylcadaverine staining, acidic vesicle formation, and the conversion of LC3BI to LC3BII. Using MM/GBSA calculations, SM-3 exhibited a stronger binding affinity (-25.09 kcal/mol) compared to Res (-18.85 kcal/mol). SM-3 also displayed greater stability during the entire simulation, maintaining lower RMSD values of 2-3 Å even after 80 ns. In summary, the introduction of methyl and methoxy functional groups on Res to create SM-3 effectively suppressed cancer spheroids and organoids formation in lung cancer cells by targeting the upstream mTOR/pAkt pathway.
Collapse
Affiliation(s)
- Zin Zin Ei
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn university, Bangkok, 10330, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Satapat Racha
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn university, Bangkok, 10330, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Interdisciplinary Program in Pharmacology, Graduate School, Chulalongkorn university, Bangkok, 10330, Thailand
| | - Preedakorn Chunhacha
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Masashi Yokoya
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Sohsuke Moriue
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Hongbin Zou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn university, Bangkok, 10330, Thailand.
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand.
- Sustainable Environment Research Institute, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
13
|
Zheng Z, Li Z, Liu X, Liu L, Zhang P, Cui Y, Ding G. Rapamycin ameliorates senescence of periodontal ligament stem cells and promotes their osteogenesis via the PI3K/AKT pathway. Int Immunopharmacol 2025; 153:114517. [PMID: 40127621 DOI: 10.1016/j.intimp.2025.114517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 02/26/2025] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
Periodontal ligament stem cells (PDLSCs) have been regarded as ideal candidates for tissue regeneration due to their excellent self-renewal and multipotent differentiation ability. Rapamycin (RAPA) is reported to play an important role in the regulation of biological properties of stem cells and a variety of physiological processes. This study investigates whether RAPA could ameliorate the senescence and accelerate the osteogenic differentiation of PDLSCs, particularly the regenerative potential in a rat calvarial bone defect model, and the underlying mechanisms involved. β-galactosidase staining, quantitative real-time polymerase chain reaction, and western blot analysis were performed to assess the effects of RAPA on senescent PDLSCs. The osteogenic differentiation ability of PDLSCs was detected by alkaline phosphatase staining and activity, Alizarin Red S staining, and gene and protein levels of osteogenesis-related markers. The underlying signaling pathways were investigated via RNA transcriptome sequencing analysis and WB tests. Calvarial bone defects in rat were treated with PDLSCs pre-incubated with or without RAPA and/or H2O2. The results showed that RAPA could enhance the osteogenic potentials of PDLSCs via PI3K/AKT signaling pathway, and reversed H2O2-induced senescence and osteogenic differentiation inhibition of PDLSCs. Moreover, calvarial defects transplanted with RAPA-treated PDLSCs showed significantly greater new bone formation compared with other groups, and also improved the H2O2-induced impairment of bone formation, whether by micro-computed tomography examination or by histological analysis. Collectively, RAPA was capable of promoting osteogenic differentiation of PDLSCs via PI3K/AKT signaling pathway in vitro, facilitating calvarial bone regeneration and reversing H2O2-induced impairment of osteogenic differentiation and cell senescence in PDLSCs.
Collapse
Affiliation(s)
- Zejun Zheng
- School of Stomatology, Shandong Second Medical University, Weifang 261053, Shandong Province, China; Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, Shandong Province, China
| | - Zekun Li
- School of Stomatology, Shandong Second Medical University, Weifang 261053, Shandong Province, China; Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, Shandong Province, China
| | - Xinjuan Liu
- School of Stomatology, Shandong Second Medical University, Weifang 261053, Shandong Province, China; Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, Shandong Province, China
| | - Luyun Liu
- School of Stomatology, Shandong Second Medical University, Weifang 261053, Shandong Province, China; Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, Shandong Province, China
| | - Ping Zhang
- School of Stomatology, Shandong Second Medical University, Weifang 261053, Shandong Province, China; Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, Shandong Province, China
| | - Yu Cui
- School of Stomatology, Shandong Second Medical University, Weifang 261053, Shandong Province, China; Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, Shandong Province, China.
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Weifang 261053, Shandong Province, China; Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, Shandong Province, China.
| |
Collapse
|
14
|
Zhou Y, Gao Y, Peng Y, Cai C, Han Y, Chen Y, Deng G, Ouyang Y, Shen H, Zeng S, Du Y, Xiao Z. QKI-induced circ_0001766 inhibits colorectal cancer progression and rapamycin resistance by miR-1203/PPP1R3C/mTOR/Myc axis. Cell Death Discov 2025; 11:192. [PMID: 40263288 PMCID: PMC12015279 DOI: 10.1038/s41420-025-02478-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/25/2025] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and remains a significant challenge due to high rates of drug resistance and limited therapeutic options. Circular RNAs (circRNAs) are increasingly recognized for their roles in CRC initiation, progression, and drug resistance. However, no circRNA-based therapies have yet entered clinical development, underscoring the need for comprehensive detection and mechanistic studies of circRNAs in CRC. Here, we identified and characterized a circular RNA, circ_0001766 (hsa_circ_0001766), through microarray analysis of CRC tissues. Our results showed that circ_0001766 is downregulated in CRC tissues and closely associated with patient survival and metastasis. Functional experiments demonstrated that circ_0001766 inhibits CRC cell proliferation, migration and invasion both in-vitro and in-vivo. Mechanistically, hypoxia downregulates Quaking (QKI), an RNA-binding protein essential for the biogenesis of circ_0001766 by binding to introns 1 and 3 of PDIA4 pre-mRNA. Reduced QKI expression under hypoxic conditions leads to decreased circ_0001766 levels in CRC. Circ_0001766 acts as a competitive endogenous RNA, sponging miR-1203 to prevent the degradation of PPP1R3C mRNA. Loss of circ_0001766 results in decreased PPP1R3C expression, leading to the activation of mTOR signaling and increased phosphorylation of Myc, which promotes CRC progression and rapamycin resistance. Our study reveals that overexpression of circ_0001766 or PPP1R3C in CRC cells inhibits the mTOR and Myc pathway, thereby resensitizing cells to rapamycin. The combination of circ_0001766 or PPP1R3C with rapamycin markedly inhibits CRC cell proliferation and induces apoptosis by reducing rapamycin-induced Myc phosphorylation. In summary, our study elucidates a critical circ_0001766/miR-1203/PPP1R3C axis that modulates CRC progression and rapamycin resistance. Our findings highlight circ_0001766 as a promising therapeutic target in CRC, providing a new avenue for enhancing the efficacy of existing treatments and overcoming drug resistance.
Collapse
Grants
- This study was supported by grants from the National Natural Science Foundation of China (No. 82373275, 81974384, 82173342 & 82203015), the China Postdoctoral Science Foundation (No.2023JJ40942), three projects from the Nature Science Foundation of Hunan Province (No.2021JJ3109, 2021JJ31048, 2023JJ40942), Nature Science Foundation of Changsha (No.73201), CSCO Cancer Research Foundation (No.Y-HR2019-0182 & Y-2019Genecast-043), the Key Research and Development Program of Hainan Province (No.ZDYF2020228 & ZDYF2020125), Natural Science Foundation (Youth Funding) of Hunan Province of China (2022JJ40458), Hunan Provincial Natural Science Foundation of China (2024JJ6662), The Youth Science Foundation of Xiangya Hospital (2023Q01) and Scientific Research Program of Hunan Provincial Health Commission (202203105261). The graphical abstract was created using BioRender (BioRender.com).This study was supported by grants from the National Natural Science Foundation of China (No. 82373275, 81974384, 82173342 & 82203015), the China Postdoctoral Science Foundation (No.2023JJ40942), three projects from the Nature Science Foundation of Hunan Province (No.2021JJ3109, 2021JJ31048, 2023JJ40942), Nature Science Foundation of Changsha (No.73201), CSCO Cancer Research Foundation (No.Y-HR2019-0182 & Y-2019Genecast-043), the Key Research and Development Program of Hainan Province (No.ZDYF2020228 & ZDYF2020125), Natural Science Foundation (Youth Funding) of Hunan Province of China (2022JJ40458), Hunan Provincial Natural Science Foundation of China (2024JJ6662), The Youth Science Foundation of Xiangya Hospital (2023Q01) and Scientific Research Program of Hunan Provincial Health Commission (202203105261). The graphical abstract was created using BioRender (BioRender.com).
Collapse
Affiliation(s)
- Yulai Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, USA
| | - Yan Gao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Yinghui Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Gongping Deng
- Department of Emergency, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Yanhong Ouyang
- Department of Emergency, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yangfeng Du
- Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China.
| | - Zemin Xiao
- Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China.
| |
Collapse
|
15
|
Tong W, Qin N, Lu T, Liu L, Liu R, Chen J, Luo N. Integrating bulk and single-cell RNA sequencing reveals SH3D21 promotes hepatocellular carcinoma progression by activating the PI3K/AKT/mTOR pathway. PLoS One 2025; 20:e0302766. [PMID: 40179068 PMCID: PMC11967960 DOI: 10.1371/journal.pone.0302766] [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: 04/11/2024] [Accepted: 02/16/2025] [Indexed: 04/05/2025] Open
Abstract
As a novel genetic biomarker, the potential role of SH3D21 in hepatocellular carcinoma remains unclear. Here, we decipher the expression and function of SH3D21 in human hepatocellular carcinoma. The expression level and clinical significance of SH3D21 in hepatocellular carcinoma patients, the relationship between SH3D21 and the features of tumor microenvironment (TME) and role of SH3D21 in promoting hepatocellular carcinoma progression were analyzed based on the bulk samples obtained from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. Single-cell sequencing samples from Gene Expression Omnibus (GEO) database were employed to verify the prediction mechanism. Additionally, different biological effects of SH3D21 on hepatocellular carcinoma cells were investigated by qRT-PCR, CCK-8 assay, colony forming assay and Western blot analysis. Bioinformatics analysis and in vitro experiments revealed that the expression level of SH3D21 was up-regulated in hepatocellular carcinoma and correlated with the poor prognosis in hepatocellular carcinoma patients. SH3D21 effectively promoted the proliferation, invasion, and migration as well as the formation of immunosuppressive microenvironment of hepatocellular carcinoma. In addition, SH3D21 can activate the PI3K/AKT/mTOR signaling pathway. SH3D21 stimulates the progression of hepatocellular carcinoma by activating the PI3K/AKT/mTOR signaling pathway, and SH3D21 can serve as a prognostic biomarker and therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Wangxia Tong
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Na Qin
- The Graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Tao Lu
- Department of hepatobiliary surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Li Liu
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Rong Liu
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jibing Chen
- Centre for Translational Medical Research in Integrative Chinese and Western Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Ning Luo
- Department of Neurology, RuiKang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
16
|
Murugan AK, Kannan S, Alzahrani AS. Immune checkpoint expression and therapeutic implications in IDH1-mutant and wild-type glioblastomas. Curr Probl Cancer 2025; 55:101182. [PMID: 39864140 DOI: 10.1016/j.currproblcancer.2025.101182] [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: 07/22/2024] [Revised: 11/17/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025]
Abstract
Programmed cell death protein 1 (PDCD1) and cluster of differentiation 274 (CD274) expression is implicated in escaping tumors from immune surveillance. Immune checkpoint inhibitors show promise in cancer therapy, yet their efficacy in glioblastomas, particularly with IDH1 mutations, remains unclear. This study analyzed two independent NGS datasets (n = 577 and n = 153) from TCGA to investigate the expression of PDCD1 and CD274 in glioblastomas and their relationship with IDH1 mutations. We used cBioPortal for mutation analysis, RNA seq for expression analysis, miRDB and miRabel for differential expression of miRNAs, and Kaplan-Meier for survival prediction. We found that 5.4% of glioblastomas harbored IDH1 mutations, correlating with improved overall survival (OS) (p = 2.196e-3). Different glioblastoma cohorts showed a diverse IDH1 mutational prevalence (4-31%). Despite this, IDH1Mu was consistently associated with better OS (p = 8.235e-5). Notably, PDCD1 and CD274 were statistically significantly highly expressed in both IDH1Wt (p < 0.0001) and IDH1Mu tumors (p < 0.0001), with higher expression linked to poorer survival outcomes (PDCD1: p = 0.009; CD274: p = 0.02). Differential co-expression analyses revealed distinct gene and miRNA profiles for IDH1Wt and IDH1Mu glioblastomas, with specific upregulation of PTEN and downregulation of MUC16 in IDH1Wt, and upregulation of PIK3R1 in IDH1Mu. Additionally, PIK3R1 and ITGB2 emerged as critical druggable targets. Our findings indicate that PDCD1 and CD274 are highly expressed irrespective of IDH1 mutation statuses, suggesting that glioblastomas could benefit from immunotherapy. Moreover, IDH1Mu glioblastomas may require a combination of PI3K/AKT/mTOR inhibitors and immunotherapy due to PIK3R1 overexpression.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia.
| | - Siddarth Kannan
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| | - Ali S Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia
| |
Collapse
|
17
|
Khayatan D, Razavi SM, Arab ZN, Nasoori H, Fouladi A, Pasha AVK, Butler AE, Karav S, Momtaz S, Abdolghaffari AH, Sahebkar A. Targeting mTOR with curcumin: therapeutic implications for complex diseases. Inflammopharmacology 2025; 33:1583-1616. [PMID: 39955697 DOI: 10.1007/s10787-025-01643-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/16/2024] [Indexed: 02/17/2025]
Abstract
The mammalian target of rapamycin (mTOR) is a crucial enzyme in regulating multiple signaling pathways in the body, including autophagy, proliferation and apoptosis. Disruption of these mTOR signaling pathways can lead to an array of abnormalities and trigger disease processes, examples being neurodegenerative conditions, cancer, obesity and diabetes. Under conditions of oxidative stress, mTOR can regulate apoptosis and autophagy, with tissue repair being favored under such circumstances. Moreover, the correlation between mTOR and other signaling pathways could play a pivotal role in the pathophysiology of numerous disorders. mTOR has a tight connection with NF-κB, Akt, PI3K, MAPK, GSK-3β, Nrf2/HO-1, JAK/STAT, CREB/BDNF, and ERK1/2 pathways, which together could play significant roles in the regulation of inflammation, apoptosis, cell survival, and oxidative stress in different body organs. Research suggests that inhibiting mTOR could be beneficial in treating metabolic, neurological and cardiovascular conditions, as well as potentially extending life expectancy. Therefore, identifying new chemicals and agents that can modulate the mTOR signaling pathway holds promise for treating and preventing these disorders. Curcumin is one such agent that has demonstrated regulatory effects on the mTOR pathway, making it an exciting alternative for reducing complications associated with complex diseases by targeting mTOR. This review aims to examine the potential of curcumin in modulating the mTOR signaling pathway and its therapeutic implications.
Collapse
Affiliation(s)
- Danial Khayatan
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Seyed Mehrad Razavi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Zahra Najafi Arab
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hadis Nasoori
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Abtin Fouladi
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aytak Vahdat Khajeh Pasha
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, Adliya, Bahrain
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Saeideh Momtaz
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
18
|
Zhang C, Xu S, Yin C, Hu S, Liu P. The role of the mTOR pathway in breast cancer stem cells (BCSCs): mechanisms and therapeutic potentials. Stem Cell Res Ther 2025; 16:156. [PMID: 40158191 PMCID: PMC11954216 DOI: 10.1186/s13287-025-04218-4] [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: 11/20/2024] [Accepted: 02/11/2025] [Indexed: 04/01/2025] Open
Abstract
Breast cancer remains the most frequently diagnosed cancer globally, exerting a profound impact on women's health and healthcare systems. Central to its pathogenesis and therapeutic resistance are breast cancer stem cells (BCSCs), which possess unique properties such as self-renewal, differentiation, and resistance to conventional therapies, contributing to tumor initiation, metastasis, and recurrence. This comprehensive review elucidates the pivotal role of the mechanistic target of rapamycin (mTOR) pathway in regulating BCSCs and its implications for breast cancer progression and treatment resistance. We explore the cellular mechanisms by which mTOR influences metastasis, metabolism, autophagy, and ferroptosis in BCSCs, highlighting its contribution to epithelial-to-mesenchymal transition (EMT), metabolic reprogramming, and survival under therapeutic stress. On a molecular level, mTOR interacts with key signaling pathways including PI3K/Akt, Notch, IGF-1R, AMPK, and TGF-β, as well as regulatory proteins and non-coding RNAs, orchestrating a complex network that sustains BCSC properties and mediates chemoresistance and radioresistance. The review further examines various therapeutic strategies targeting the mTOR pathway in BCSCs, encompassing selective PI3K/Akt/mTOR inhibitors, monoclonal antibodies, natural products, and innovative approaches such as nanoparticle-mediated drug delivery. Clinical trials investigating mTOR inhibitors like sirolimus and combination therapies with agents such as everolimus and trastuzumab are discussed, underscoring their potential in eradicating BCSCs and improving patient outcomes. Additionally, natural compounds and repurposed drugs offer promising adjunctive therapies by modulating mTOR activity and targeting BCSC-specific vulnerabilities. In conclusion, targeting the mTOR pathway presents a viable and promising avenue for enhancing breast cancer treatment efficacy by effectively eliminating BCSCs, reducing tumor recurrence, and improving overall patient survival. Continued research and clinical validation of mTOR-targeted therapies are essential to translate these insights into effective clinical interventions, ultimately advancing personalized cancer management and therapeutic outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shu Xu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Chuanzheng Yin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Shaobo Hu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
| |
Collapse
|
19
|
Teng C, Chen JW, Shen LS, Chen S, Chen GQ. Research advances in natural sesquiterpene lactones: overcoming cancer drug resistance through modulation of key signaling pathways. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:13. [PMID: 40201307 PMCID: PMC11977367 DOI: 10.20517/cdr.2024.178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 04/10/2025]
Abstract
Cancer remains a significant global health challenge, with current chemotherapeutic strategies frequently limited by the emergence of resistance. In this context, natural compounds with the potential to overcome resistance have garnered considerable attention. Among these, sesquiterpene lactones, primarily derived from plants in the Asteraceae family, stand out for their potential anticancer properties. This review specifically focuses on five key signaling pathways: PI3K/Akt/mTOR, NF-κB, Wnt/β-catenin, MAPK/ERK, and STAT3, which play central roles in the mechanisms of cancer resistance. For each of these pathways, we detail their involvement in both cancer development and the emergence of drug resistance. Additionally, we investigate how sesquiterpene lactones modulate these pathways to overcome resistance across diverse cancer types. These insights highlight the potential of sesquiterpene lactones to drive the advancement of novel therapies that can effectively combat both cancer progression and drug resistance.
Collapse
Affiliation(s)
- Chi Teng
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, Guangdong, China
- Authors contributed equally
| | - Jia-Wen Chen
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, Guangdong, China
- Authors contributed equally
| | - Li-Sha Shen
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Sibao Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, Guangdong, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Guo-Qing Chen
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, Guangdong, China
| |
Collapse
|
20
|
Tanwar SS, Dwivedi S, Khan S, Sharma S. Cardiomyopathies and a brief insight into DOX-induced cardiomyopathy. Egypt Heart J 2025; 77:29. [PMID: 40064787 PMCID: PMC11893974 DOI: 10.1186/s43044-025-00628-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Cardiomyopathy is a heterogeneous group of myocardial disorders characterized by structural and functional abnormalities of the heart muscle. It is classified into primary (genetic, mixed, or acquired) and secondary categories, resulting in various phenotypes including dilated, hypertrophic, and restrictive patterns. Hypertrophic cardiomyopathy, the most common primary form, can cause exertional dyspnea, presyncope, and sudden cardiac death. Dilated cardiomyopathy typically presents with heart failure symptoms, while restrictive cardiomyopathy is rarer and often associated with systemic diseases. Diagnosis involves a comprehensive evaluation including history, physical examination, electrocardiography, and echocardiography. Treatment options range from pharmacotherapy and lifestyle modifications to implantable cardioverter-defibrillators and heart transplantation in refractory cases. MAIN BODY Anthracyclines, particularly doxorubicin, have emerged as crucial components in cancer treatment, demonstrating significant antitumor activity across various malignancies. These drugs have become standard in numerous chemotherapy regimens, improving patient outcomes. However, their use is associated with severe cardiotoxicity, including cardiomyopathy and heart failure. The mechanisms of anthracycline action and toxicity are complex, involving DNA damage, iron-mediated free radical production, and disruption of cardiovascular homeostasis. Doxorubicin-induced cardiomyopathy (DIC) is a severe complication of cancer treatment with a poor prognosis and limited effective treatments. The pathophysiology of DIC involves multiple mechanisms, including oxidative stress, inflammation, mitochondrial damage, and calcium homeostasis disorder. Despite extensive research, no effective treatment for established DIC is currently available. Dexrazoxane is the only FDA-approved protective agent, but it has limitations. Recent studies have explored various potential therapeutic approaches, including natural drugs, endogenous substances, new dosage forms, and herbal medicines. However, the lack of experimental models incorporating pre-existing cancer limits the understanding of DIC pathophysiology and treatment efficacy. CONCLUSION Cardiomyopathy, whether primary or secondary, poses a significant clinical challenge due to its varying etiologies and poor prognosis in advanced stages. Anthracycline-induced cardiomyopathy is a severe complication of chemotherapy, with doxorubicin being a notable contributor. Despite advancements in cancer therapies, the cardiotoxic effects of anthracyclines necessitate further investigation into effective preventive strategies and therapeutic interventions to improve patient outcomes.
Collapse
Affiliation(s)
| | - Sumeet Dwivedi
- Acropolis Institute of Pharmaceutical Education and Research, Indore, India
| | - Sheema Khan
- The University of Texas Rio Grande Valley, Edinburg, US
| | - Seema Sharma
- Shri Vaishnav Vidyapeeth Vishwadvidyalaya, Indore, India.
| |
Collapse
|
21
|
Xiong N, Wang Y, Jiang J. USP5 Promotes Head and Neck Squamous Cell Carcinoma Progression via mTOR Signaling Pathway. Cancer Med 2025; 14:e70752. [PMID: 40066708 PMCID: PMC11894462 DOI: 10.1002/cam4.70752] [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: 12/02/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a highly aggressive malignancy characterized by limited prognostic markers and treatment options, contributing to high mortality rates. While Ubiquitin-specific peptidase 5 (USP5) has been implicated in various cancers, its role in HNSCC remains poorly understood. AIMS This study aims to investigate the role of USP5 in the progression of HNSCC and explore its potential as both a prognostic biomarker and a therapeutic target. MATERIALS & METHODS This work utilized single-cell transcriptomic analysis with the Scissor algorithm to identify distinct epithelial subpopulations, particularly focusing on the Stress subpopulation that exhibited significant upregulation of USP5. Validation was conducted using tissue microarray (TMA) analysis and immunohistochemistry (IHC) to compare USP5 expression levels in HNSCC tissues versus adjacent normal tissues. Furthermore, RNA interference (RNAi) experiments were performed to knock down USP5 expression, assessing its effects on tumor cell behavior, including proliferation, migration, and invasion, as well as the regulation of mTORC1 and NF-κB signaling pathways. RESULTS This study revealed that the Stress subpopulation, characterized by USP5 upregulation, was associated with enhanced tumor cell proliferation, migration, and invasion. TMA and IHC analyses confirmed that USP5 expression was significantly higher in HNSCC tissues compared to normal tissues, correlating with poor patient prognosis. Additionally, RNAi-mediated knockdown of USP5 led to reduced tumor cell activities and downregulation of the mTORC1 and NF-κB signaling pathways. DISCUSSION The findings suggest that USP5 plays a critical role in driving HNSCC progression. Its overexpression in aggressive tumor subpopulations and association with poor clinical outcomes highlight its potential utility as both a prognostic biomarker and a therapeutic target. The observed effects on cell behavior and oncogenic signaling pathways provide mechanistic insights into how USP5 for HNSCC therapy. CONCLUSIONS This study establishes USP5 as a key driver of HNSCC progression, underscoring its potential role in prognosis and therapy. Targeting USP5 may offer novel treatment strategies for HNSCC, addressing the urgent need for effective therapeutic interventions in this aggressive malignancy.
Collapse
Affiliation(s)
- Ni Xiong
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yue Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of StomatologyGuanghua School of Stomatology, Sun Yat‐Sen UniversityGuangzhouChina
| | - Junhong Jiang
- Department of StomatologyJiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| |
Collapse
|
22
|
Kwon MJ, Raut PK, Jang JH, Chun KS. Isoliquiritigenin Induces Apoptosis via ROS-Mediated Inhibition of p38/mTOR/STAT3 Pathway in Human Melanoma Cells. Biomol Ther (Seoul) 2025; 33:378-387. [PMID: 39933948 PMCID: PMC11893486 DOI: 10.4062/biomolther.2024.118] [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: 07/21/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 02/13/2025] Open
Abstract
Isoliquiritigenin (ISL), a phenolic compound derived from licorice, exhibits various biological activities, including anti-inflammatory, anti-viral, anti-tumor, and antioxidant effects. However, the molecular mechanisms underlying its anti-cancer effects are not well understood in SK-MEL-28 melanoma cells. Melanoma, a highly aggressive and treatment-resistant cancer, remains a significant health challenge. This study investigates the anti-cancer effects of ISL, focusing on identifying reactive oxygen species (ROS)-mediated apoptosis mechanisms on SK-MEL-28 melanoma cells. Our results show that ISL treatment induces apoptosis in SK-MEL-28 cells, as evidenced by the cleavage of caspase-9, -7, -3, and PARP. ISL increased Bax expression, decreased Bcl-2 expression, and promoted cytochrome C release into the cytosol. ISL also reduced the expression of cell cycle markers, including cyclin D1, D3, and survivin. Notably, ISL treatment markedly increased intracellular ROS levels and pretreatment with N-acetyl cysteine, a ROS scavenger, abrogated the ISL-induced inhibition of the p38/mTOR/STAT3 pathway and prevented apoptosis. Moreover, ISL significantly diminished the constitutive phosphorylation of mTOR and STAT3 in SK-MEL-28 cells by blocking the phosphorylation of p38 MAPK, an upstream kinase of mTOR. Pharmacological inhibition of mTOR attenuated the STAT3 signaling, indicating that mTOR acts as an upstream kinase of STAT3 in these cells. Collectively, these findings demonstrate that ISL inhibits SK-MEL-28 cell growth by downregulating cell survival proteins and inducing apoptosis through ROS generation.
Collapse
Affiliation(s)
- Mi Jeong Kwon
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Pawan Kumar Raut
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Jeong-Hoon Jang
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
23
|
Jurca AA, Jurca AD, Petchesi CD, Bembea D, Jurca CM, Severin E, Jurca S, Vesa CM. Tuberous Sclerosis Complex: New Insights into Pathogenesis and Therapeutic Breakthroughs. Life (Basel) 2025; 15:368. [PMID: 40141713 PMCID: PMC11944049 DOI: 10.3390/life15030368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Tuberous sclerosis complex (TSC) is a rare, autosomal dominant genetic disorder caused by mutations in the TSC1 and TSC2 genes, which disrupt the regulation of the mammalian target of rapamycin (mTOR) pathway, a critical regulator of cellular growth. The disorder presents as a multisystem condition, with benign tumors (hamartomas) developing in organs such as the brain, skin, heart, kidneys, and lungs, leading to significant clinical variability and impact on quality of life. This review aims to summarize recent advances in the understanding of TSC pathogenesis and clinical variability and evaluate the therapeutic breakthroughs in targeted treatments. Methods: A narrative review was conducted using various available databases. We applied objective evaluation metrics, such as the impact factor of the journals and the citation count, to assess the quality of the studies. Results: Targeted therapies, particularly mTOR inhibitors (mTORis), have shown efficacy in reducing hamartoma size, improving neuropsychiatric symptoms, and enhancing patient outcomes. Despite these advances, variability in disease expression poses challenges in diagnosis and individualized management strategies. Conclusions: Challenges such as early diagnosis, optimizing long-term outcomes, and addressing residual unmet needs remain critical. Future research should prioritize precision medicine approaches and patient-centered care models within centers of expertise to improve treatment efficacy and quality of life for individuals with TSC.
Collapse
Affiliation(s)
- Aurora Alexandra Jurca
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania;
| | - Alexandru Daniel Jurca
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 1 December Sq., 410081 Oradea, Romania; (C.D.P.); (D.B.); (C.M.J.); (C.M.V.)
| | - Codruta Diana Petchesi
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 1 December Sq., 410081 Oradea, Romania; (C.D.P.); (D.B.); (C.M.J.); (C.M.V.)
- Regional Center of Medical Genetics Bihor, County Emergency Clinical Hospital Oradea (Part of ERN THACA), 410469 Oradea, Romania
| | - Dan Bembea
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 1 December Sq., 410081 Oradea, Romania; (C.D.P.); (D.B.); (C.M.J.); (C.M.V.)
| | - Claudia Maria Jurca
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 1 December Sq., 410081 Oradea, Romania; (C.D.P.); (D.B.); (C.M.J.); (C.M.V.)
- Regional Center of Medical Genetics Bihor, County Emergency Clinical Hospital Oradea (Part of ERN THACA), 410469 Oradea, Romania
| | - Emilia Severin
- Department of Genetics, University of Medicine and Pharmacy “Carol Davila”, Dionisie Lupu Street, Number 37, District 2, 020021 Bucharest, Romania
| | - Sanziana Jurca
- Faculty of Medicine and Pharmacy, University of Oradea, December Sq., 410081 Oradea, Romania;
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 1 December Sq., 410081 Oradea, Romania; (C.D.P.); (D.B.); (C.M.J.); (C.M.V.)
| |
Collapse
|
24
|
Pradhan PM, Lee YH, Jang S, Yi HK. Synergistic anti-cancer effects of metformin and cisplatin on YD-9 oral squamous carcinoma cells via AMPK pathway. J Appl Oral Sci 2025; 33:e20240385. [PMID: 40008711 DOI: 10.1590/1678-7757-2024-0385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/07/2025] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVE This study evaluated whether hypoglycemic drug metformin enhances the anti-cancer effects of cisplatin in YD-9 cells. METHODOLOGY YD-9 cells, derived from oral mucosal squamous cell carcinoma of oral mucosa, were used to assess the combined effects of metformin and cisplatin by means of MTT assay, live and dead cell staining, and colony formation assays to evaluate cell viability and proliferation. Reactive oxygen species level was measured using a Muse cell analyzer. Apoptosis, epithelial-mesenchymal transition, and related molecular pathways were analyzed by western blot. Wound healing assays and Transwell migration assays examined cell migration, whereas monophosphate-activated protein kinase inhibitor Compound C, was utilized to investigate the AMPK pathway. RESULTS Sequential treatment of YD-9 cells with metformin and cisplatin resulted in decreased cell viability and proliferation, increased ROS levels, and elevated apoptosis compared with the individual drugs. Moreover, the treatment inhibited EMT, wound healing, and cell migration. These results correlated with increased AMPK phosphorylation, a key regulator of cellular energy homeostasis. Introduction of Compound C pre-treatment upregulated N-cadherin and α-smooth muscle actin along with enhanced cell migration. CONCLUSION This study found synergism in anti-cancer effects between metformin and cisplatin. Additionally, introduction of Compound C confirmed that EMT inhibition is AMPK dependent. These findings indicate the potential use of metformin as an adjunct drug in anti-cancer treatments, warranting further investigation.
Collapse
Affiliation(s)
- Paras Man Pradhan
- Jeonbuk National University, Institute of Oral Bioscience, School of Dentistry, Department of Oral Biochemistry, Jeonju, Korea
| | - Young-Hee Lee
- Jeonbuk National University, Institute of Oral Bioscience, School of Dentistry, Department of Oral Biochemistry, Jeonju, Korea
| | - Sungil Jang
- Jeonbuk National University, Institute of Oral Bioscience, School of Dentistry, Department of Oral Biochemistry, Jeonju, Korea
| | - Ho-Keun Yi
- Jeonbuk National University, Institute of Oral Bioscience, School of Dentistry, Department of Oral Biochemistry, Jeonju, Korea
| |
Collapse
|
25
|
Luo M, Jin T, Fang Y, Chen F, Zhu L, Bai J, Ding J. Signaling Pathways Involved in Acute Pancreatitis. J Inflamm Res 2025; 18:2287-2303. [PMID: 40230438 PMCID: PMC11995411 DOI: 10.2147/jir.s485804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 01/25/2025] [Indexed: 04/16/2025] Open
Abstract
Acute pancreatitis (AP) is a common digestive emergency with high morbidity and mortality. Over the past decade, significant progress has been made in understanding the mechanisms of AP, including oxidative stress, disruptions in calcium homeostasis, endoplasmic reticulum stress, inflammatory responses, and various forms of cell death. This review provides an overview of the typical signaling pathways involved and proposes the latest clinical translation prospects. These strategies are important for the early management of AP, preventing multi-organ injury, and improving the overall prognosis of the disease.
Collapse
Affiliation(s)
- Mengchen Luo
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Ting Jin
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Yi Fang
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Feng Chen
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Lujian Zhu
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Jin Ding
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| |
Collapse
|
26
|
Deng Z, Guo J, Zhu Z, Qing Q, Wan D, Lei P, Liu Q, Huang B. The effect and mechanism of atorvastatin regulating PI3K-Akt-mTOR pathway on radiosensitivity of hepatocellular carcinoma cells. Toxicol Res (Camb) 2025; 14:tfae202. [PMID: 40012843 PMCID: PMC11851483 DOI: 10.1093/toxres/tfae202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/24/2024] [Accepted: 02/22/2025] [Indexed: 02/28/2025] Open
Abstract
Radiation therapy is an important method to treat liver cancer, but because of the strong DNA repair ability of liver cancer cells, even after receiving high doses of radiation still can not get satisfactory results. Atorvastatin (ATO) is a lipophilic and tissue-selective inhibitor of HMG-CoA reductase whose anticancer effects have been validated in various cells, but its effect on the radiation sensitivity of hepatocellular carcinoma cells remains unclear. Therefore, Therefore, this study explored the radiosensitivity of ATO and its possible mechanism by pretreating HepG2 with ATO and collecting HepG2 cells after irradiation. It was found that atorvastatin can not only affect the survival of liver cancer cells when used alone, but also enhance the radiation sensitivity of HepG2 cells. The study found that ATO significantly exacerbated the inhibitory effects of IR on the growth, proliferation, and migration of HepG2 cells. Measurement of ROS, SOD, GPx, and MDA levels indicated that ATO enhanced IR-induced oxidative stress, further promoted the decrease of Mitochondrial Membrane Potential, increased the rate of apoptosis in HepG2, upregulating pro-apoptotic proteins Bax and Cleaved-Caspase 3, and downregulating anti-apoptotic proteins Bcl-2. Western blot analysis showed that the PI3K-Akt-mTOR pathway was inhibited, leading to the activation of cytotoxic autophagy in HepG2 and an increase in the expression of the LC-3II protein. In summary, ATO, in combination with IR, enhances the oxidative stress response of HepG2 induced by IR, promotes autophagy by inhibiting the PI3K-Akt-mTOR pathway, and thereby potentially enhances the radiosensitivity of HepG2 as a pharmacological intervention.
Collapse
Affiliation(s)
- Zhengzheng Deng
- College of Public Health, Hengyang Medical School, University of South China, 28 West Changsheng Road, Hengyang, Hunan 421000, P.R. China
| | - Jinjing Guo
- College of Public Health, Hengyang Medical School, University of South China, 28 West Changsheng Road, Hengyang, Hunan 421000, P.R. China
| | - Zihao Zhu
- College of Public Health, Hengyang Medical School, University of South China, 28 West Changsheng Road, Hengyang, Hunan 421000, P.R. China
| | - Qiancheng Qing
- College of Public Health, Hengyang Medical School, University of South China, 28 West Changsheng Road, Hengyang, Hunan 421000, P.R. China
| | - Dangting Wan
- College of Public Health, Hengyang Medical School, University of South China, 28 West Changsheng Road, Hengyang, Hunan 421000, P.R. China
- Guangdong Maoming Health Vocational College, No. 1 Anle East Road, Maoming City, Guangdong 525000, P.R. China
| | - Pengyuan Lei
- College of Public Health, Hengyang Medical School, University of South China, 28 West Changsheng Road, Hengyang, Hunan 421000, P.R. China
| | - Qi Liu
- College of Public Health, Hengyang Medical School, University of South China, 28 West Changsheng Road, Hengyang, Hunan 421000, P.R. China
| | - Bo Huang
- College of Public Health, Hengyang Medical School, University of South China, 28 West Changsheng Road, Hengyang, Hunan 421000, P.R. China
| |
Collapse
|
27
|
Liu Y, Zhang W, Jang H, Nussinov R. mTOR Variants Activation Discovers PI3K-like Cryptic Pocket, Expanding Allosteric, Mutant-Selective Inhibitor Designs. J Chem Inf Model 2025; 65:966-980. [PMID: 39792006 PMCID: PMC12091942 DOI: 10.1021/acs.jcim.4c02022] [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: 11/04/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
mTOR plays a crucial role in PI3K/AKT/mTOR signaling. We hypothesized that mTOR activation mechanisms driving oncogenesis can advise effective therapeutic designs. To test this, we combined cancer genomic analysis with extensive molecular dynamics simulations of mTOR oncogenic variants. We observed that conformational changes within mTOR kinase domain are associated with multiple mutational activation events. The mutations disturb the α-packing formed by the kαAL, kα3, kα9, kα9b, and kα10 helices in the kinase domain, creating cryptic pocket. Its opening correlates with opening of the catalytic cleft, including active site residues realignment, favoring catalysis. The cryptic pocket created by disrupted α-packing coincides with the allosteric pocket in PI3Kα can be harmoniously fitted by the PI3Kα allosteric inhibitor RLY-2608, suggesting that analogous drugs designed based on RLY-2608 can restore the packed α-structure, resulting in mTOR inactive conformation. Our results exemplify that knowledge of detailed kinase activation mechanisms can inform innovative allosteric inhibitor development.
Collapse
Affiliation(s)
- Yonglan Liu
- Cancer
Innovation Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Wengang Zhang
- Cancer
Innovation Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Hyunbum Jang
- Cancer
Innovation Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
- Computational
Structural Biology Section, Frederick National
Laboratory for Cancer Research, Frederick, Maryland 21702, United States
| | - Ruth Nussinov
- Cancer
Innovation Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
- Computational
Structural Biology Section, Frederick National
Laboratory for Cancer Research, Frederick, Maryland 21702, United States
- Department
of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
28
|
Li Q, Li Y, Zhou T, Zhang Y, Li H, Yuan F, Bi Y. FBXW7 metabolic reprogramming inhibits the development of colon cancer by down-regulating the activity of arginine/mToR pathways. PLoS One 2025; 20:e0317294. [PMID: 39823500 PMCID: PMC11741656 DOI: 10.1371/journal.pone.0317294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025] Open
Abstract
FBXW7 is a tumor suppressor gene that regulates metabolism and is associated with the onset and progression of colorectal cancer (CRC)), however, the precise mechanism whereby FBXW7 participates in the metabolic reprogramming of CRC remains unclear. Here, the research aims to reveal the association between the expression of FBXW7 and clinical variables and to investigate the molecular mechanism by which FBXW7 plays a critical role in the development of CRC. The clinical importance of FBXW7 in CRC was determined by immunohistochemistry. Non-targeted metabolomics was utilized to explore the role of FBXW7 in the metabolic regulation of CRC. Low expression of FBXW7 was associated with poor prognosis in individuals with CRC, both at the mRNA and protein levels. FBXW7 over-expression inhibited CRC cell growth, colony formation, migration, and invasion. Non-targeted metabolomics unveiled that FBXW7 over-expression directly caused the deprivation of arginine which led to downmodulation of mTOR signaling pathway; meanwhile, FBXW7-related metabolites were primarily concentrated in the mTOR signaling pathway. In summary, the research identified a novel mechanism of action of FBXW7 in CRC. The research findings provide a theoretical foundation for the prognostic prediction and therapeutic planning of CRC based on metabolic reprogramming.
Collapse
Affiliation(s)
- Qing Li
- Center of Gene Sequencing, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, P. R. China
| | - Yan Li
- Department of Epidemiology, Academy of Medical Sciences, School of Public Health, Shanxi Medical University, Taiyuan, P. R. China
| | - Tong Zhou
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yong Zhang
- Endoscopic Center of Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, P. R. China
| | - Huiyu Li
- Department of General Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Fajia Yuan
- Shanxi Jinzhong Health School, Jinzhong, P. R. China
| | - Yanghui Bi
- Center of Gene Sequencing, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, P. R. China
| |
Collapse
|
29
|
Kumar D, Kanchan R, Chaturvedi NK. Targeting protein synthesis pathways in MYC-amplified medulloblastoma. Discov Oncol 2025; 16:23. [PMID: 39779613 PMCID: PMC11711608 DOI: 10.1007/s12672-025-01761-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
MYC is one of the most deregulated oncogenic transcription factors in human cancers. MYC amplification/or overexpression is most common in Group 3 medulloblastoma and is positively associated with poor prognosis. MYC is known to regulate the transcription of major components of protein synthesis (translation) machinery, leading to promoted rates of protein synthesis and tumorigenesis. MTOR signaling-driven deregulated protein synthesis is widespread in various cancers, including medulloblastoma, which can promote the stabilization of MYC. Indeed, our previous studies demonstrate that the key components of protein synthesis machinery, including mTOR signaling and MYC targets, are overexpressed and activated in MYC-amplified medulloblastoma, confirming MYC-dependent addiction of enhanced protein synthesis in medulloblastoma. Further, targeting this enhanced protein synthesis pathway with combined inhibition of MYC transcription and mTOR translation by small-molecule inhibitors, demonstrates preclinical synergistic anti-tumor potential against MYC-driven medulloblastoma in vitro and in vivo. Thus, inhibiting enhanced protein synthesis by targeting the MYC indirectly and mTOR pathways together may present a highly appropriate strategy for treating MYC-driven medulloblastoma and other MYC-addicted cancers. Evidence strongly proposes that MYC/mTOR-driven tumorigenic signaling can predominantly control the translational machinery to elicit cooperative effects on increased cell proliferation, cell cycle progression, and genome dysregulation as a mechanism of cancer initiation. Several small molecule inhibitors of targeting MYC indirectly and mTOR signaling have been developed and used clinically with immunosuppressants and chemotherapy in multiple cancers. Only a few of them have been investigated as treatments for medulloblastoma and other pediatric tumors. This review explores concurrent targeting of MYC and mTOR signaling against MYC-driven medulloblastoma. Based on existing evidence, targeting of MYC and mTOR pathways together produces functional synergy that could be the basis for effective therapies against medulloblastoma.
Collapse
Affiliation(s)
- Devendra Kumar
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 986395, USA
| | - Ranjana Kanchan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nagendra K Chaturvedi
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 986395, USA.
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
30
|
Feng X, Wang Z, Cen M, Zheng Z, Wang B, Zhao Z, Zhong Z, Zou Y, Lv Q, Li S, Huang L, Huang H, Qiu X. Deciphering potential molecular mechanisms in clear cell renal cell carcinoma based on the ubiquitin-conjugating enzyme E2 related genes: Identifying UBE2C correlates to infiltration of regulatory T cells. Biofactors 2025; 51:e2143. [PMID: 39614426 DOI: 10.1002/biof.2143] [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: 09/19/2024] [Accepted: 10/28/2024] [Indexed: 12/01/2024]
Abstract
Renal clear cell carcinoma (ccRCC) is a highly aggressive and common form of kidney cancer, with limited treatment options for advanced stages. Recent studies have highlighted the importance of the ubiquitin-proteasome system in tumor progression, particularly the role of ubiquitin-conjugating enzyme E2 (UBE2) family members. However, the prognostic significance of UBE2-related genes (UBE2RGs) in ccRCC remains unclear. In this study, bulk RNA-sequencing and single-cell RNA-sequencing data from ccRCC patients were retrieved from the Cancer Genome Atlas and Gene Expression Omnibus databases. Differential expression analysis was performed to identify UBE2RGs associated with ccRCC. A combination of 10 machine learning methods was applied to develop an optimal prognostic model, and its predictive performance was evaluated using area under the curve (AUC) values for 1-, 3-, and 5-year overall survival (OS) in both training and validation cohorts. Functional enrichment analyses of gene ontology and Kyoto Encyclopedia of Genes and Genomes were conducted to explore the biological pathways involved. Correlation analysis was conducted to investigate the association between the risk score and tumor mutational burden (TMB) and immune cell infiltration. Immunotherapy and chemotherapy sensitivity were assessed by immunophenoscore and tumor immune, dysfunction, and exclusion scores to identify potential predictive significance. In vitro, knockdown of the key gene UBE2C in 786-O cells by specific small interfering RNA to validate its impact on apoptosis, migration, cell cycle, migration, invasion of tumor cells, and induction of regulatory T cells (Tregs). Analysis of sc-RNA revealed that UBE2 activity was significantly upregulated in malignant cells, suggesting its role in tumor progression. A three-gene prognostic model comprising UBE2C, UBE2D3, and UBE2T was constructed by Lasoo Cox regression and demonstrated robust predictive accuracy, with AUC values of 0.745, 0.766, and 0.771 for 1-, 3-, and 5-year survival, respectively. The model was validated as an independent prognostic factor in ccRCC. Patients in the high-risk group had a worse prognosis, higher TMB scores, and low responsiveness to immunotherapy. Additionally, immune infiltration and chemotherapy sensitivity analyses revealed that UBE2RGs are associated with various immune cells and drugs, suggesting that UBE2RGs could be a potential therapeutic target for ccRCC. In vitro experiments confirmed that the reduction of UBE2C led to an increase in apoptosis rate, as well as a decrease in tumor cell invasion and metastasis abilities. Additionally, si-UBE2C cells reduced the release of the cytokine Transforming Growth Factor-beta 1 (TGF-β1), leading to a decreased ratio of Tregs in the co-culture system. This study presents a novel three-gene prognostic model based on UBE2RGs that demonstrates significant predictive value for OS, immunotherapy, and chemotherapy in ccRCC patients. The findings underscore the potential of UBE2 family members as biomarkers and therapeutic targets in ccRCC, warranting further investigation in prospective clinical trials.
Collapse
Affiliation(s)
- Xiaoqiang Feng
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Zhenwei Wang
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Meini Cen
- Department of Rehabilitation Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zongtai Zheng
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Bangqi Wang
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Zongxiang Zhao
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Zhihui Zhong
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Yesong Zou
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Qian Lv
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Shiyu Li
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Hai Huang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Urology, Ruijin Hospital Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofu Qiu
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Shahrokhi Nejad S, Razi S, Rezaei N. The role of AMPK in pancreatic cancer: from carcinogenesis to treatment. Clin Transl Oncol 2025; 27:70-82. [PMID: 38926257 DOI: 10.1007/s12094-024-03572-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Pancreatic cancer has doubled over the previous two decades. Routine therapies are becoming incredibly resistant and failing to compensate for the burden caused by this aggressive neoplasm. As genetic susceptibility has always been a highlighted concern for this disease, identifying the molecular pathways involved in the survival and function of pancreatic cancer cells provides insight into its variant etiologies, one of which is the role of AMPK. This regulating factor of cell metabolism is crucial in the homeostasis and growth of the cell. Herein, we review the possible role of AMPK in pancreatic cancer while considering its leading effects on glycolysis and autophagy. Then, we assess the probable therapeutic agents that have resulted from the suggested pathways. Studying the underlying genetic changes in pancreatic cancer provides a chance to detect and treat patients suffering from advanced stages of the disease, and those who have given up their hope on conventional therapies can gain an opportunity to combat this cancer.
Collapse
Affiliation(s)
- Shahrzad Shahrokhi Nejad
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, 14194, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
32
|
Mandal SK, Samanta SK. A Perspective of PI3K/AKT/mTOR Pathway Inhibitors to Overcome Drug-resistance in Breast Cancer Therapy. Curr Med Chem 2025; 32:1865-1873. [PMID: 39171586 DOI: 10.2174/0109298673327425240815065221] [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: 05/01/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024]
Abstract
The heterogeneous disease, breast cancer (BC), is a frequently detected cancer today, including hormone receptor-positive (HR+), human epidermal growth factor receptor-2-positive (HER2+), and triple-negative (ER-, PR-, HER2-) BC. Advanced endocrine therapies could improve about 85% HR+ BC patient survival. Still, 20% - 30% of cases of endocrine therapy resistance are observed. For all kinds of breast cancer, drug resistance is a common and dangerous phenomenon, comprised of two types: de novo resistance and acquired resistance (prolonged exposure). According to recent works of literature, the PI3K/AKT/mTOR pathway has become an emerging target for overcoming drug resistance in BC therapy due to its close association with tumour growth and resistance from current therapies. Activation of the PI3K/AKT/mTOR pathway was found to promote multidrug resistance by elevating drugs' outflow. The first orally active PI3K inhibitor, Alpelisib (BYL-719) in fulvestrant combination, was approved for treating HR+/ HER2- metastatic BC. Therefore, utilizing PI3K/mTOR/AKT inhibitors in combination with currently available strategies could be an optimistic approach to overcoming drug resistance and resensitizing drug-resistant tumor cells of BC. Here, in this perspective, BC cancer therapies related to drug resistance, the involvement of PI3K/AKT/mTOR pathway in drug resistance and multi-drug resistance, and the role of PI3K/AKT/mTOR inhibitors in getting rid of drug resistance have been illuminated.
Collapse
Affiliation(s)
- Sudip Kumar Mandal
- Dr. B.C. Roy College of Pharmacy and Allied Health Sciences, Bidhannagar, Durgapur, 713212, West Bengal, India
| | - Samir Kumar Samanta
- Dr. B.C. Roy College of Pharmacy and Allied Health Sciences, Bidhannagar, Durgapur, 713212, West Bengal, India
| |
Collapse
|
33
|
Wang Z, Zhang Y, Xue Y, Huang W, Zhang H. EIF2S2 transcriptionally upregulates HIF1α to promote gastric cancer progression via activating PI3K/AKT/mTOR pathway. Carcinogenesis 2024; 45:916-927. [PMID: 39046731 DOI: 10.1093/carcin/bgae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 06/24/2024] [Accepted: 07/22/2024] [Indexed: 07/25/2024] Open
Abstract
Eukaryotic translation initiation factor 2 subunit beta (EIF2S2) is a protein that controls protein synthesis under various stress conditions and is abnormally expressed in several cancers. However, there is limited insight regarding the expression and molecular role of EIF2S2 in gastric cancer. In this study, we identified the overexpression of EIF2S2 in gastric cancer by immunohistochemical staining and found a positive correlation between EIF2S2 expression and shorter overall survival and disease-free survival. Functionally, we revealed that EIF2S2 knockdown suppressed gastric cancer cell proliferation and migration, induced cell apoptosis, and caused G2 phase cell arrest. Additionally, EIF2S2 is essential for in vivo tumor formation. Mechanistically, we demonstrated that EIF2S2 transcriptionally regulated hypoxia-inducible factor-1 alpha (HIF1α) expression by NRF1. The promoting role of EIF2S2 in malignant behaviors of gastric cancer cells depended on HIF1α expression. Furthermore, the PI3K/AKT/mTOR signaling was activated upon EIF2S2 overexpression in gastric cancer. Collectively, EIF2S2 exacerbates gastric cancer progression via targeting HIF1α, providing a fundamental basis for considering EIF2S2 as a potential therapeutic target for gastric cancer patients.
Collapse
Affiliation(s)
- Zhiyong Wang
- Department of Gastrointestinal Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Road, Wuhan 430022, Hubei Province, China
| | - Yingyi Zhang
- Department of Oncology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, China
| | - Yingwei Xue
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang Province, China
| | - Wei Huang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, No. 150 Jimo Road, Shanghai 200120, China
| | - Hongfeng Zhang
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang Province, China
| |
Collapse
|
34
|
Yin L, Mao L, Yin R, Lv C, Shi X, Yue C, Chen Y, Lu C, Wu Z, Xu K, Cao W. ACE Loss Drives Renal Cell Carcinoma Growth and Invasion by Modulating AKT-FOXO1. Biologics 2024; 18:397-412. [PMID: 39717370 PMCID: PMC11665188 DOI: 10.2147/btt.s485178] [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: 08/22/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Purpose Emerging literature links the role of the renin-angiotensin-aldosterone system (RAAS) to the progression of cancers. However, the function of RAAS has not been verified in Clear-cell renal cell carcinoma (ccRCC). Methods ACE expression in ccRCC tissues was determined using RT-PCR, Western blot, and immunohistochemistry staining. The clinical significance of ACE was evaluated through Cox regression analysis. To assess the impact of ACE expression on ccRCC cell growth, metastasis, and glucose activity, CCK-8 assays, transwell assays, Seahorse detection, and xenograft models were utilized. The mechanisms of ACE and its upstream and downstream regulatory factors were investigated using RNA-seq, chromatin immunoprecipitation (ChIP), and luciferase reporter assays. Results RAAS-related gene Angiotensin-Converting Enzyme (ACE) was significantly under expressed in ccRCC cells and tissues. High ACE expression was positively associated with a favorable prognosis in ccRCC patients. Functional studies showed that ACE overexpression suppressed ccRCC cell line OS-RC-2 and A498 growth, metastasis, and glycolysis activities, while its knockdown had the opposite effect. Mechanistically, ACE inhibited ccRCC progression and epithelial-mesenchymal transition (EMT) by disrupting the AKT-FOXO1 signaling pathway. Furthermore, we provide evidence that ACE could enhance everolimus (approved agent for ccRCC) antitumor effect and ACE expression is transcriptionally regulated by ZBTB26. Conclusion Our findings investigated the roles and mechanisms of ACE in ccRCC. ACE inhibits the growth and metastasis of ccRCC cells in vitro and in vivo by promoting FOXO1 expression, which is the downstream target of PI3K-AKT pathway. Thus, this research suggests that ACE may be a promising target for new therapeutic strategy in ccRCC.
Collapse
Affiliation(s)
- Lei Yin
- Department of Urology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, People’s Republic of China
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Department of Urology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, People’s Republic of China
| | - Lixin Mao
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Rui Yin
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
| | - Chengxun Lv
- Department of Urology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, People’s Republic of China
| | - Xiaokai Shi
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Chuang Yue
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Yin Chen
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Chao Lu
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Zonglin Wu
- Department of Urology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, People’s Republic of China
| | - Kai Xu
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Wei Cao
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| |
Collapse
|
35
|
Li JF, Jin L, Ma H, Suo J, Yang R, Yang XP. 1.25(OH)2D3 decreases PCNA and mTOR expression and alleviates renal injury in Thy-1 nephritis rat model. PLoS One 2024; 19:e0311000. [PMID: 39636964 PMCID: PMC11620346 DOI: 10.1371/journal.pone.0311000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/10/2024] [Indexed: 12/07/2024] Open
Abstract
OBJECTIVE This study investigated the role and mechanisms of 1.25(OH)2D3 in proliferative glomerulonephritis and its effect on the regulation of mesangial cells. METHODS Sixty male SD rats were randomly divided into four groups: control (CG), nephritis (NG), nephritis + 1.25(OH)2D3(NVG), and nephritis + 1.25(OH)2D3+ rapamycin (NVRG) (n = 15 per group). Three rats from each group were sacrificed on days 1, 3, 7, 14, and 21 after intervention. Urine samples were collected over 24 hours on day 0 to measure urinary protein excretion. Renal tissue samples were stained with HE and PAS to evaluate the extent of renal injury, while immunohistochemistry was employed to quantify PCNA and mTOR expression in the renal tissues. RESULTS Compared to the NG, mesangial cell proliferation in the renal tissues was significantly reduced in the NVG and NVRG at all time points (all p<0.05). PCNA expressionwas significantly higher in the NG compared to the CG (p < 0.05) and significantly lower in the NVG and NVRG (p < 0.05). mTOR expression was also significantly increased in the NG compared to the CG, with a significant reduction observed in the NVG and NVRG compared to the NG. CONCLUSION Our findings demonstrate that 1.25(OH)2D3significantly inhibits the proliferation of glomerular mesangial cells in rats. Additionally, mTOR protein is involved in the regulation of glomerular mesangial cells by 1.25(OH)2D3. These results further elucidate the molecular mechanism by which 1.25(OH)2D3alleviates renal injury in glomerulonephritis.
Collapse
Affiliation(s)
- Jian-feng Li
- Department of Nephrology, The First People’s Hospital of Nanyang City, Nanyang, Henan Province, China
| | - Lei Jin
- Department of Rheumatology, Immunology & Allergy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Ma
- Department of Gastrointestinal Surgery, The First People’s Hospital of Nanyang City, Nanyang, Henan Province, China
| | - Jie Suo
- Department of Nephrology, The First People’s Hospital of Aksu City, Aksu, Xin jiang Province, China
| | - Rui Yang
- Department of Nephrology, The First Affiliated Hospital of Shi he zi University, Shihezi City, Xin jiang Province, China
| | - Xiao-ping Yang
- Department of Nephrology, The First Affiliated Hospital of Shi he zi University, Shihezi City, Xin jiang Province, China
| |
Collapse
|
36
|
Zheng L, Wei N, Farooqi AA, Zhang Y, Blundell R, Liu X, Xu Y, Lin X. Recent progress of protein kinase inhibitors derived from marine peptides for developing anticancer agents. Cell Signal 2024; 124:111411. [PMID: 39277091 DOI: 10.1016/j.cellsig.2024.111411] [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: 07/13/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Protein kinases, mediating their biological function via their catalytic activity, play important role in cell development, including cell proliferation, migration, angiogenesis and survival. Over the years, protein kinase inhibitors have been developed as an important class of anticancer agents clinically. However, the off-targeting and drug resistance of protein kinase inhibitors limit their efficiency. Anticancer peptides derived from marine organisms represent a novel class of bioactive substances, and some of the peptides exhibit anticancer effect via inhibiting protein kinases. In this mini review, the recent progress of anticancer peptides targeting protein kinases from marine sources are presented. Marine peptides inhibiting resistant cancer cells by targeting novel domains of protein kinases are highlighted. The challenges and prospects of developing marine peptides as anticancer agents are also discussed.
Collapse
Affiliation(s)
- Lanhong Zheng
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, PR China.
| | - Ning Wei
- Department of Oncology and Cancer Therapeutics Program, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, Pakistan.
| | - Yan Zhang
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, PR China.
| | - Renald Blundell
- Center for Molecular Medicine and Biobanking, University of Malta, Imsida MSD2080, Malta.
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Peking Union Medical College, Beijing 100050, China.
| | - Yixin Xu
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, PR China.
| | - Xiukun Lin
- College of Marine Sci, Beibu Gulf University, 12 Binhai Rd., Qinzhou 535001, PR China.
| |
Collapse
|
37
|
Millar MW, Najar RA, Slavin SA, Shadab M, Tahir I, Mahamed Z, Lin X, Abe JI, Wright TW, Dean DA, Fazal F, Rahman A. MTOR maintains endothelial cell integrity to limit lung vascular injury. J Biol Chem 2024; 300:107952. [PMID: 39510184 PMCID: PMC11664419 DOI: 10.1016/j.jbc.2024.107952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 09/21/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
The functional and structural integrity of the endothelium is essential for vascular homeostasis. Loss of barrier function in quiescent and migratory capacity in proliferative endothelium causes exuberant vascular permeability, a cardinal feature of many inflammatory diseases including acute lung injury (ALI). However, the signals governing these fundamental endothelial cell (EC) functions are poorly understood. Here, we identify mechanistic target of rapamycin (MTOR) as an important link in preserving the barrier integrity and migratory/angiogenic responses in EC and preventing lung vascular injury and mortality in mice. Knockdown of MTOR in EC altered cell morphology, impaired proliferation and migration, and increased endocytosis of cell surface vascular endothelial (VE)-cadherin leading to disrupted barrier function. MTOR-depleted EC also exhibited reduced VE-cadherin and vascular endothelial growth factor receptor-2 (VEGFR2) levels mediated in part by autophagy. Similarly, lungs from mice with EC-specific MTOR deficiency displayed spontaneous vascular leakage marked by decreased VE-cadherin and VEGFR2 levels, indicating that MTOR deficiency in EC is sufficient to disrupt lung vascular integrity and may be a key pathogenic mechanism of ALI. Indeed, MTOR as well as VEGFR2 and VE-cadherin levels were markedly reduced in injured mouse lungs or EC. Importantly, EC-targeted gene transfer of MTOR complementary DNA, either prophylactically or therapeutically, mitigated inflammatory lung injury, and improved lung function and survival in mouse models of ALI. These findings reveal an essential role of MTOR in maintaining EC function, identify loss of endothelial MTOR as a key mechanism of lung vascular injury, and show the therapeutic potential of EC-targeted MTOR expression in combating ALI and mortality in mice.
Collapse
Affiliation(s)
- Michelle Warren Millar
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Rauf A Najar
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Spencer A Slavin
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Mohammad Shadab
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Imran Tahir
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Zahra Mahamed
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Xin Lin
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Terry W Wright
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - David A Dean
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Fabeha Fazal
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Arshad Rahman
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| |
Collapse
|
38
|
Hu HF, Fu JY, Han L, Gao GB, Zhang WX, Yu SM, Li N, Li YJ, Lu YF, Ding XF, Pan YL, Wang Y, He QY. The Antipsychotic Drug Aripiprazole Suppresses Colorectal Cancer by Targeting LAMP2a to Induce RNH1/miR-99a/mTOR-Mediated Autophagy and Apoptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409498. [PMID: 39513392 DOI: 10.1002/advs.202409498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/24/2024] [Indexed: 11/15/2024]
Abstract
The mammalian target of rapamycin (mTOR) is a critical signaling hub for sustaining cancer survival. Targeting mTOR and inducing autophagic cell death downstream of it represent promising therapeutic strategies for cancer prevention. A US Food and Drug Administration-approved drug library containing 616 small molecules is used to screen anticancer drugs against colorectal cancer (CRC) cells that rely on mTOR. This led to the identification of an antipsychotic drug aripiprazole, which significantly induced mTOR inhibition and autophagic apoptosis in CRC, in vitro and in vivo. The use of drug affinity response target stability identified lysosome-associated membrane protein 2A (LAMP2a) as a direct target of aripiprazole. LAMP2a-deficient CRC cells are refractory to aripiprazole. High LAMP2a expression is associated with poor survival of patients with CRC and negatively correlated with expression of ribonuclease inhibitor 1 (RNH1), which is later confirmed as a novel substrate of LAMP2a. Mechanistically, aripiprazole bound to the Lys401-His404 of LAMP2a and repressed its activity, subsequently inactivating RNH1/miR-99a/mTOR signaling and inducing autophagy-mediated apoptosis, thereby suppressing tumorigenesis. Liposome-mediated delivery of aripiprazole in combination with fluorouracil elicited superior therapeutic benefits in CRC, as compared to single treatments, thereby highlighting that aripiprazole may be repurposed as a novel therapeutic agent for CRC treatment.
Collapse
Affiliation(s)
- Hui-Fang Hu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
- The First Affiliated Hospital of Jinan University and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Jia-Ying Fu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Lei Han
- Institute of Biomedicine and National Engineering Research Center of Genetic Medicine and Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, National Engineering Research Centre of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Gui-Bin Gao
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Wei-Xia Zhang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Si-Ming Yu
- Institute of Biomedicine and National Engineering Research Center of Genetic Medicine and Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, National Engineering Research Centre of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Nan Li
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yang-Jia Li
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yi-Fan Lu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xiao-Feng Ding
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yun-Long Pan
- The First Affiliated Hospital of Jinan University and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Yang Wang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
- The First Affiliated Hospital of Jinan University and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
39
|
Feng Y, Jiang Q, Ma X, Sun H, Chai Y, Li X, Wang Z, Feng F. Photosensitizing metal-organic framework nanoparticles combined with tumor-sensitization strategies can enhance the phototherapeutic effect upon medullary thyroid carcinoma. Biochim Biophys Acta Gen Subj 2024; 1868:130725. [PMID: 39433228 DOI: 10.1016/j.bbagen.2024.130725] [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: 07/08/2024] [Revised: 09/23/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
Photodynamic therapy (PDT) utilizing metal-organic frameworks (MOFs) has developed as a new and efficacious treatment for malignant tumors located on the surface of the human body. In order to achieve more effective PDT treatment outcomes, the traditional method has been to increase the intensity of the laser irradiation, but this approach can easily lead to tissue burns. In this study, we developed a new type of nanoparticle, F68-PKI@PCN224, aims to achieve effective PDT upon medullary thyroid carcinoma (MTC) which is an uncommon form of thyroid cancer that originates in the parafollicular cells of the thyroid and the therapeutic outlook for patients with MTC remains unsatisfactory. F68-PKI@PCN224 combines the antitumor features of PDT with mammalian target of rapamycin (mTOR) inhibitor PKI-587 (PKI). The tumor sensitization, slow release, and pH response features of F68-PKI@PCN224 was demonstrated by a series of in vitro and in vivo experiments / assays. F68-PKI@PCN224 achieved the long-term activation and slow releasing of PKI and TCPP in MTC tumor tissues. During the process of generating PDT effects, F68-PKI@PCN224 enhanced the tumor's sensitivity to PDT, direct laser irradiation of MTC cells or subcutaneous tumor tissues. As a result, low-dose phototherapy achieves a higher anti-tumor effect upon F68-PKI@PCN224 compared with TCPP. This study reveals the synergistic effect between tumor sensitization by mTOR inhibitor and PDT and initially unveils the mechanism of action of these nanoparticles.
Collapse
Affiliation(s)
- Yingqi Feng
- Beijing Key Laboratory for Green Catalysis and Separation and Department of Chemical Engineering, College of Materials Science & Engineering, Beijing University of Technology, Beijing 100124, PR China; Clinical Laboratory, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100039, China
| | - Qiyu Jiang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Institute of Infectious Diseases, Beijing 100039, China
| | - Xue Ma
- The First affiliated Hospital of Jinzhou Medical University, Jinzhou 121017, Liaoning Province, China
| | - Huiwei Sun
- Department of Infectious Diseases, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Institute of Infectious Diseases, Beijing 100039, China
| | - Yantao Chai
- Clinical Laboratory, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100039, China
| | - Xiaojuan Li
- Department of Infectious Diseases, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Institute of Infectious Diseases, Beijing 100039, China
| | - Zhijie Wang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Institute of Infectious Diseases, Beijing 100039, China
| | - Fan Feng
- Clinical Laboratory, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100039, China.
| |
Collapse
|
40
|
Vozgirdaite D, Hervé-Aubert K, Uzbekov R, Chourpa I, Allard-Vannier E. Design, optimization, characterization, and in vitro evaluation of metformin-loaded liposomes for triple negative breast cancer treatment. J Liposome Res 2024; 34:547-561. [PMID: 38459750 DOI: 10.1080/08982104.2024.2321528] [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: 10/06/2023] [Revised: 01/22/2024] [Accepted: 02/16/2024] [Indexed: 03/10/2024]
Abstract
Recently, metformin (Met) has shown to have antineoplastic properties in cancer treatment by improving hypoxic tumor conditions, and causing reduction in the synthesis of biomolecules, which are vital for cancer growth. However, as an orally administered drug, Met has low bioavailability and rapid renal clearance. Thus, the goal of this study was to vectorize Met inside liposomes in the context of triple negative breast cancer (TNBC), which currently lacks treatment options when compared to other types of breast cancer. Vectorization of Met inside liposomes was done using Bangham method by implementing double design of experiment methodology to increase Met drug loading (minimum-run resolution V characterization design and Box-Behnken design), as it is generally extremely low for hydrophilic molecules. Optimization of Met-loaded liposome synthesis was successfully achieved with drug loading of 190 mg/g (19% w/w). The optimal Met-liposomes were 170 nm in diameter with low PdI (< 0.1) and negative surface charge (-20 mV), exhibiting sustained Met release at pH 7.4. The liposomal Met delivery system was stable over several months, and successfully reduced TNBC cell proliferation due to the encapsulated drug. This study is one the first reports addressing liposome formulation through thin-film hydration using two design of experiment methods aiming to increase drug loading of Met.
Collapse
Affiliation(s)
- Daiva Vozgirdaite
- UPR 4301 CBM, CNRS, NMNS department, University of Tours, Tours, France
| | | | - Rustem Uzbekov
- Laboratoire Biologie Cellulaire et Microscopie Electronique, Faculty of Medicine, University of Tours, Tours, France
- Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow, Russia
| | - Igor Chourpa
- UPR 4301 CBM, CNRS, NMNS department, University of Tours, Tours, France
| | | |
Collapse
|
41
|
Li C, Du X, Zhang H, Liu S. Knockdown of ribosomal protein L22-like 1 arrests the cell cycle and promotes apoptosis in colorectal cancer. Cytojournal 2024; 21:45. [PMID: 39737125 PMCID: PMC11683392 DOI: 10.25259/cytojournal_29_2024] [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: 03/11/2024] [Accepted: 09/27/2024] [Indexed: 01/01/2025] Open
Abstract
Objective Colorectal cancer (CRC) remains a remarkable challenge despite considerable advancements in its treatment, due to its high recurrence rate, metastasis, drug resistance, and heterogeneity. Molecular targets that can effectively inhibit CRC growth must be identified to address these challenges. Therefore, we aim to reveal the regulatory effect of ribosomal protein L22-like 1 (RPL22L1) on the proliferation and apoptosis of CRC cells and its potential mechanism. Material and Methods We detected the expression of RPL22L1 from the Cancer Genome Atlas, Gene Expression Omnibus and UALCAN databases. The effects of RPL22L1 on CRC growth and migration were determined by knocking down RPL22L1 in human CRC cell lines and those on the cell cycle and apoptosis using flow cytometry. The influence of RPL22L1 knockdown on xenograft tumor growth was verified in vivo. The potential RPL22L1 mechanisms in promoting cancer were predicted with RNA sequencing (RNAseq). The molecular mechanism of enhanced apoptosis and cell cycle arrest in RPL22L1 knockdown was revealed using real-time reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) and Western blotting. Results The present study reveals a considerable upregulation of RPL22L1 expression in CRC as well as in diverse tumor tissues, and most cells within the CRC tumor microenvironment (TME) demonstrate RPL22L1 expression. Notably, this elevated expression level of RPL22L1 exhibits a strong association with an unfavorable prognosis among patients diagnosed with CRC (P < 0.05). Furthermore, the association between RPL22L1 expression and the CRC TME index did not exhibit statistical significance (P > 0.05). However, RPL22L1 knockdown experiments revealed a substantial suppression of growth and migratory capacities in CRC cells RKO and HCT116 (P < 0.05). Flow cytometry analysis exhibited that on RPL22L1 knockdown, a remarkable arrest of the G1 and S phases of the cell cycle (P < 0.05) occurred. In addition, a remarkable elevation in the level of cell apoptosis was observed (P < 0.001). RNAseq exhibited that cell cycle, DNA replication, and mechanistic target of rapamycin (mTOR) complex 1pathway were inhibited after RPL22L1 knockdown, whereas the apoptosis pathway was activated (P < 0.05). Validation through RT-qPCR and western blot analysis also corroborated the downregulation of P70S6K, MCM3, MCM7, GADD45B, WEE1, and MKI67 expression levels, following RPL22L1 knockdown (P < 0.05). Consequent rescue experiments offered supportive evidence, indicating the involvement of the mTOR pathway in mediating the influence of RPL22L1 on the promotion of cell cycle progression. Moreover, in vivo assays involving tumor-bearing mice exhibited that diminished RPL22L1 levels led to arrested CRC growth (P < 0.05). Conclusion These findings support RPL22L1 as a possible prognostic and therapeutic target in CRC, providing novel insights into the development of anticancer medications.
Collapse
Affiliation(s)
- Chunming Li
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, College of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Xinna Du
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, College of Basic Medicine, Jiamusi University, Jiamusi, China
- Department of Physiology and Biochemistry, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Hu Zhang
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, College of Basic Medicine, Jiamusi University, Jiamusi, China
- Department of Physiology and Biochemistry, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Shuang Liu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, College of Basic Medicine, Jiamusi University, Jiamusi, China
| |
Collapse
|
42
|
Han Y, Deng X, Chen H, Chen J, Xu W, Liu L. Succinylation modification-mediated upregulation of Sp1 promotes hepatocellular carcinoma cell proliferation. Discov Oncol 2024; 15:660. [PMID: 39548054 PMCID: PMC11568111 DOI: 10.1007/s12672-024-01533-9] [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: 05/29/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the most prevalent malignant tumors globally, characterized by high incidence and mortality rates. Despite ongoing research, the underlying molecular mechanisms of HCC development are not yet fully understood. Utilizing bioinformatic analysis, real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR), and Western blot assays, we identified that the expression of specificity protein 1 (Sp1) was significantly elevated in HCC cells compared to normal cells. Knockdown of the Sp1 gene led to a marked reduction in the viability and clonogenic potential of HCC cells. Further investigation revealed that the succinylation level of Sp1 was also increased in HCC cells. The upregulation of Sp1 expression was attributed to its succinylation, mediated by KAT2A, with lysine (K)562 identified as the succinylation site. Additionally, KAT2A and Sp1 were found to influence the upregulation of mTOR phosphorylation. Collectively, these findings suggest that KAT2A-promoted succinylation of Sp1 enhances the proliferative capacity of HCC cells by activating the mTOR pathway, providing a theoretical foundation for potential therapeutic strategies against HCC.
Collapse
Affiliation(s)
- Yehong Han
- General surgery, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, No.453, Stadium Road, Hangzhou, 310007, Zhejiang, China
| | - Xueqin Deng
- General surgery, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, No.453, Stadium Road, Hangzhou, 310007, Zhejiang, China
| | - Haixia Chen
- General surgery, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, No.453, Stadium Road, Hangzhou, 310007, Zhejiang, China
| | - Jie Chen
- General surgery, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, No.453, Stadium Road, Hangzhou, 310007, Zhejiang, China
| | - Wei Xu
- General surgery, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, No.453, Stadium Road, Hangzhou, 310007, Zhejiang, China
| | - Lanqin Liu
- General Neurology Department, Hangzhou TCM Hospital affiliated to Zhejiang Chinese Medical University, No.453, Stadium Road, Hangzhou, 310007, Zhejiang, China.
| |
Collapse
|
43
|
Liao C, He Y, Luo X, Deng G. Ferroptosis: insight into the treatment of hepatocellular carcinoma. Cancer Cell Int 2024; 24:376. [PMID: 39538215 PMCID: PMC11562710 DOI: 10.1186/s12935-024-03559-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignances in the world, with high morbidity and mortality. Due to the hidden onset of symptoms, there are huge obstacles in early diagnosis, recurrence, metastasis and drug resistance. Although great strides have been made in the treatment of HCC, effective treatment options are still limited and achieving longer survival for patients remains urgent. Ferroptosis is a novel type of programmed cell death that is mainly caused by iron-dependent oxidative damage. With further investigations, ferroptosis has been proved to be associated with the occurrence and development of various tumors. This article reviews the regulatory mechanism and signal transduction pathways of ferroptosis, investigates the complex relationship between autophagy, sorafenib resistance and immunotherapy with ferroptosis involved in HCC, providing new ideas and directions for the treatment of HCC.
Collapse
Affiliation(s)
- Chuanjie Liao
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, China
| | - Youwu He
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, China
| | - Xinning Luo
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, China
| | - Ganlu Deng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, China.
| |
Collapse
|
44
|
Wen JY, Fang YY, Li DM, Tang YL, Huang HQ, Liu LM, Zeng JH, Dang YW, Pan YF, Zeng DT, Huang WJ, Chen G, Li H. A Comprehensive Analysis of LYAR in Colorectal Cancer: Prognostic Marker and Therapeutic Target. Cancer Biother Radiopharm 2024; 39:673-689. [PMID: 39159060 DOI: 10.1089/cbr.2023.0181] [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] [Indexed: 08/21/2024] Open
Abstract
Background: Colorectal cancer (CRC) is a major global health challenge with a need for new biomarkers and therapeutic targets. This work investigated the biological mechanisms and clinical value of Ly1 antibody reactive (LYAR) in CRC. Methods: We analyzed LYAR mRNA expression across multiple public databases, including genotype-tissue expression, gene expression omnibus, Oncomine, and the cancer genome atlas, alongside in-house immunohistochemical data to evaluate LYAR protein expression in CRC and non-CRC colorectal tissues. Gene set enrichment analysis (GSEA) was used to elucidate LYAR's biological functions, and its impact on the tumor immune microenvironment was assessed using CIBERSORT, ESTIMATE, and single-cell RNA sequencing techniques. In addition, LYAR's association with clinicopathological features and patient prognosis was explored, and its influence on drug sensitivity was investigated using the Connectivity Map database. Results: LYAR was significantly upregulated in CRC tissues compared with non-CRC colorectal counterparts, associated with altered immune cell composition and enhanced RNA processing, splicing, and cell cycle regulation. High LYAR expression correlated with poor disease-free and overall survival, underscoring its prognostic value. GSEA revealed LYAR's involvement in critical cellular processes and pathways, including DNA repair, cell cycle, and mTORC1 signaling. Correlation analysis identified genes positively and negatively associated with LYAR, leading to the discovery of temsirolimus and WYE-354, mTOR inhibitors, as potential therapeutic agents for CRC. Furthermore, LYAR expression predicted increased sensitivity to cetuximab in RAS wild-type metastatic CRC, indicating its utility as a biomarker for treatment responsiveness. Conclusions: LYAR's upregulation in CRC highlights its potential as a biomarker for prognosis and therapeutic targeting, offering insights into CRC pathology and suggesting new avenues for treatment optimization.
Collapse
Affiliation(s)
- Jia-Ying Wen
- Department of Radiotherapy, The Second Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Ye-Ying Fang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Dong-Ming Li
- Department of Pathology, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Yu-Lu Tang
- Department of Pathology, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - He-Qing Huang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Li-Min Liu
- Department of Toxicology, College of Pharmacy, Guangxi Medical University, Nanning, P.R. China
| | - Jiang-Hui Zeng
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangxi Medical University/Nanning Second People's Hospital, Nanning, P. R. China
| | - Yi-Wu Dang
- Department of Pathology, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Yan-Fang Pan
- Department of Pathology, Hospital of Guangxi Liugang Medical Co., LTD./Guangxi Liuzhou Dingshun Forensic Expert Institute, Liuzhou, P.R. China
| | - Da-Tong Zeng
- Department of Pathology, Redcross Hospital of Yulin city, Yulin, P.R. China
| | - Wei-Jian Huang
- Department of Pathology, Redcross Hospital of Yulin city, Yulin, P.R. China
| | - Gang Chen
- Department of Pathology, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Hui Li
- Department of Colorectal & Anal Surgery, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| |
Collapse
|
45
|
Zhang Y, Wang Y, Weng J, Chen J, Zheng Y, Xia Y, Huang Z, Zhao L, Chen X, Tang H, Huang Y. Pyroptosis-related gene GSDMC indicates poor prognosis and promotes tumor progression by activating the AKT/mTOR pathway in lung squamous cell carcinoma. Mol Carcinog 2024; 63:2218-2236. [PMID: 39136610 DOI: 10.1002/mc.23805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/23/2024] [Accepted: 07/29/2024] [Indexed: 10/11/2024]
Abstract
Lung squamous cell carcinoma (LUSC) is one of the most common malignant tumors of the respiratory. Pyroptosis plays an essential role in cancer, but there is limited research investigating pyroptosis in LUSC. In this study, pyroptosis-related genes were observed to have extensive multiomics alterations in LUSC through analysis of the TCGA database. Utilizing machine learning for selection and verifying expression levels, GSDMC was chosen as the critical gene for further experiments. Our research found that GSDMC is overexpressed in LUSC tissues and cells, and is associated with poor prognosis. Knockdown of GSDMC in LUSC inhibits cell proliferation, invasion, metastasis, chemotherapeutic sensitivity, and reduced tumor formation in nude mice, accompanied by downregulation of proliferative and EMT-related protein expression. However, these effects were counteracted in cells where GSDMC is overexpressed. Mechanistically, the oncogenic role of GSDMC is primarily achieved through the activation of the AKT/mTOR pathway, and this effect can be significantly reversed by rapamycin. Finally, SMAD4's interaction with the promoter region of GSDMC results in the suppression of GSDMC expression. In summary, our study through bioinformatics and experimental approaches not only proves that SMAD4 regulates the protumorigenic role of GSDMC through transcriptional targeting, but also indicates the possibility of developing the SMAD4/GSDMC/AKT/mTOR signaling axis as a potential biomarker and treatment target for LUSC.
Collapse
Affiliation(s)
- Yi Zhang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Yuzhi Wang
- Department of Laboratory Medicine, Deyang People's Hospital, Deyang, Sichuan, China
| | - Jiamiao Weng
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Jianlin Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Yue Zheng
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Yu Xia
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
- Integrated Chinese and Western Medicine College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Zhixin Huang
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
- Integrated Chinese and Western Medicine College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Lilan Zhao
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Department of General Thoracic Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiongfeng Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Department of Scientific Research, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Haijun Tang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fuzhou, Fujian, China
| | - Yi Huang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fuzhou, Fujian, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, China
- Central Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, China
| |
Collapse
|
46
|
Wang J, Wen Y, Zhang Y, Wang Z, Jiang Y, Dai C, Wu L, Leng D, He S, Bo X. An interpretable artificial intelligence framework for designing synthetic lethality-based anti-cancer combination therapies. J Adv Res 2024; 65:329-343. [PMID: 38043609 PMCID: PMC11519055 DOI: 10.1016/j.jare.2023.11.035] [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: 09/05/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/05/2023] Open
Abstract
INTRODUCTION Synthetic lethality (SL) provides an opportunity to leverage different genetic interactions when designing synergistic combination therapies. To further explore SL-based combination therapies for cancer treatment, it is important to identify and mechanistically characterize more SL interactions. Artificial intelligence (AI) methods have recently been proposed for SL prediction, but the results of these models are often not interpretable such that deriving the underlying mechanism can be challenging. OBJECTIVES This study aims to develop an interpretable AI framework for SL prediction and subsequently utilize it to design SL-based synergistic combination therapies. METHODS We propose a knowledge and data dual-driven AI framework for SL prediction (KDDSL). Specifically, we use gene knowledge related to the SL mechanism to guide the construction of the model and develop a method to identify the most relevant gene knowledge for the predicted results. RESULTS Experimental and literature-based validation confirmed a good balance between predictive and interpretable ability when using KDDSL. Moreover, we demonstrated that KDDSL could help to discover promising drug combinations and clarify associated biological processes, such as the combination of MDM2 and CDK9 inhibitors, which exhibited significant anti-cancer effects in vitro and in vivo. CONCLUSION These data underscore the potential of KDDSL to guide SL-based combination therapy design. There is a need for biomedicine-focused AI strategies to combine rational biological knowledge with developed models.
Collapse
Affiliation(s)
- Jing Wang
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yuqi Wen
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Yixin Zhang
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Zhongming Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Yuyang Jiang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Chong Dai
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lianlian Wu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Dongjin Leng
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Song He
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| | - Xiaochen Bo
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| |
Collapse
|
47
|
Zhong L, Wu J, Zhou B, Kang J, Wang X, Ye F, Lin X. ALYREF recruits ELAVL1 to promote colorectal tumorigenesis via facilitating RNA m5C recognition and nuclear export. NPJ Precis Oncol 2024; 8:243. [PMID: 39455812 PMCID: PMC11512073 DOI: 10.1038/s41698-024-00737-0] [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: 04/30/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
ALYREF can recognize 5-methylcytosine (m5C) decoration throughout RNAs to regulate RNA metabolism. However, its implications in cancer and precise regulatory mechanisms remain largely elusive. Here, we demonstrated that ALYREF supported colorectal cancer (CRC) growth and migration. Integrated analysis of ALYREF-RIP-Bis-seq and transcriptome profiles identified ribosomal protein S6 kinase B2 (RPS6KB2) and regulatory-associated protein of mTOR (RPTOR) as ALYREF's possible downstream effectors. Mechanistically, ALYREF formed a complex with ELAV like RNA binding protein 1 (ELAVL1) to cooperatively promote m5C recognition and nuclear export of the two mRNAs. Moreover, ALYREF protein was highly expressed in tumor tissues of CRC patients, which predicted their poor prognosis. E2F transcription factor 6 (E2F6)-mediated transactivation gave a molecular insight into ALYREF overexpression. Collectively, ALYREF recruits ELAVL1 to collaboratively facilitate m5C recognition and nuclear export of RPS6KB2 and RPTOR transcripts for colorectal tumorigenesis, providing RNA m5C methylation as promising therapeutic targets and prognostic biomarkers for CRC.
Collapse
Affiliation(s)
- Longhua Zhong
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, P. R. China
| | - Jingxun Wu
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, P. R. China
| | - Bingqian Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, P. R. China
| | - Jiapeng Kang
- Department of Medical Oncology, Zhangzhou Municipal Hospital, Zhangzhou Municipal Hospital Affiliated of Fujian Medical University, Zhangzhou, P. R. China
| | - Xicheng Wang
- Cancer Medical Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China.
| | - Feng Ye
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, P. R. China.
- Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, China.
| | - Xiaoting Lin
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, P. R. China.
- Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, China.
| |
Collapse
|
48
|
Ameer SF, Mohamed MY, Elzubair QA, Sharif EAM, Ibrahim WN. Curcumin as a novel therapeutic candidate for cancer: can this natural compound revolutionize cancer treatment? Front Oncol 2024; 14:1438040. [PMID: 39507759 PMCID: PMC11537944 DOI: 10.3389/fonc.2024.1438040] [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: 05/24/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024] Open
Abstract
Cancer remains one of the leading causes of death worldwide. Despite advances in medical treatments, current therapeutic strategies, including radiotherapy, chemotherapy, targeted therapy, and surgical resection, have not significantly reduced the global incidence and mortality rates of cancer. Oncologists face considerable challenges in devising effective treatment plans due to the adverse side effects associated with standard therapies. Therefore, there is an urgent need for more effective and well-tolerated cancer treatments. Curcumin, a naturally occurring compound, has garnered significant attention for its diverse biological properties. Both preclinical studies and clinical trials have highlighted curcumin's potential in cancer treatment, demonstrating its ability to inhibit the proliferation of various cancer cell types through multiple cellular and molecular pathways. This paper examines the antineoplastic properties, and the therapeutic mechanisms including cell signalling pathways targeted by curcumin that are implicated in cancer development and explores the challenges in advancing curcumin as a viable anticancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
49
|
Shilovsky GA. p62: Intersection of Antioxidant Defense and Autophagy Pathways. Mol Biol 2024; 58:822-835. [DOI: 10.1134/s0026893324700390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/20/2024] [Accepted: 05/07/2024] [Indexed: 01/05/2025]
|
50
|
Deng Z, Qing Q, Huang B. A bibliometric analysis of the application of the PI3K-AKT-mTOR signaling pathway in cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7255-7272. [PMID: 38709265 DOI: 10.1007/s00210-024-03112-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
PI3K-AKT-mTOR plays as important role in the growth, metabolism, proliferation, and migration of cancer cells, and in apoptosis, autophagy, inflammation, and angiogenesis in cancer. In this study, the aim was to comprehensively review the current research landscape regarding the PI3K-AKT-mTOR pathway in cancer, using bibliometrics to analyze research hotspots, and provide ideas for future research directions. Literature published on the topic between January 2006 and May 2023 was retrieved from the Web of Science core database, and key information and a visualization map were analyzed using CiteSpace and VOSviewer. A total of 5800 articles from 95 countries/regions were collected, including from China and the USA. The number of publications on the topic increased year on year. The major research institution was the University of Texas MD Anderson Cancer Center. Oncotarget and Clinical Cancer Research were the most prevalent journals in the field. Of 26,621 authors, R Kurzrock published the most articles, and J Engelman was cited most frequently. "A549 cell," "first line treatment," "first in human phase I," and "inhibitor" were the keywords of emerging research hotspots. Inhibitors of the PI3K-AKT-mTOR pathway and their use in clinical therapeutic strategies for cancer were the main topics in the field, and future research should also focus on PI3K-AKT-mTOR pathway inhibitors. This study is the first to comprehensively summarize trends and development s in research into the PI3K-AKT-mTOR pathway in cancer. The information that was obtained clarified recent research frontiers and directions, providing references for scholars of cancer management.
Collapse
Affiliation(s)
- Zhengzheng Deng
- School of Public Health, University of South China, Hengyang, 421001, Hunan Province, China
| | - Qiancheng Qing
- School of Public Health, University of South China, Hengyang, 421001, Hunan Province, China
| | - Bo Huang
- School of Public Health, University of South China, Hengyang, 421001, Hunan Province, China.
| |
Collapse
|