251
|
Zhang H, Hu H, Li Y, Wang J, Ma L. A ferrocene-based hydrogel as flexible electrochemical biosensor for oxidative stress detection and antioxidation treatment. Biosens Bioelectron 2024; 248:115997. [PMID: 38183792 DOI: 10.1016/j.bios.2023.115997] [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: 11/13/2023] [Revised: 12/27/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Real-time sensing of reactive oxygen species (ROS) and timely scavenging of excessive ROS in physiological environments are critically important in the diagnosis and prevention of ROS-related diseases. To solve the mismatch problem between conventional rigid ROS biosensors and biological tissues in terms of both modulus and composition, here, we present a flexible ferrocene-based hydrogel biosensor designed for oxidative stress detection and antioxidation treatment. The hydrogel was fabricated through a supramolecular assembly of ferrocene-grafted polyethylenimine (PEI-Fc), sodium alginate (SA), and polyvinyl alcohol (PVA). Multiple non-covalent interactions, including electrostatic interactions between PEI-Fc and SA, hydrophobic interactions and π-π stacking among ferrocene groups, and the PVA crystalline domain, synergistically improve the mechanical properties of the PVA/SA/PEI-Fc hydrogel. The flexible PVA/SA/PEI-Fc hydrogel biosensor exhibited a broad detection range for hydrogen peroxide (H2O2), from 0 to 120 μM, using the differential pulse voltammetry method. Furthermore, the hydrogel demonstrated effective ROS scavenging and oxygen generation performance, desirable biocompatibility, and satisfactory antibacterial activity, making it suitable for biological interfaces. In vitro studies revealed that the PVA/SA/PEI-Fc hydrogel could monitor H2O2 concentration in the proximity of inflammatory cells, and effectively scavenge ROS to protect cells from oxidative stress damage. This all-in-one multifunctional hydrogel, integrating both sensing and treatment functions, holds great promise for clinical applications in the diagnosis and management of ROS-related diseases.
Collapse
Affiliation(s)
- Haiqi Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Hongtao Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yan Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jinze Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lie Ma
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
252
|
Sharma A, Verwilst P, Li M, Ma D, Singh N, Yoo J, Kim Y, Yang Y, Zhu JH, Huang H, Hu XL, He XP, Zeng L, James TD, Peng X, Sessler JL, Kim JS. Theranostic Fluorescent Probes. Chem Rev 2024; 124:2699-2804. [PMID: 38422393 PMCID: PMC11132561 DOI: 10.1021/acs.chemrev.3c00778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The ability to gain spatiotemporal information, and in some cases achieve spatiotemporal control, in the context of drug delivery makes theranostic fluorescent probes an attractive and intensely investigated research topic. This interest is reflected in the steep rise in publications on the topic that have appeared over the past decade. Theranostic fluorescent probes, in their various incarnations, generally comprise a fluorophore linked to a masked drug, in which the drug is released as the result of certain stimuli, with both intrinsic and extrinsic stimuli being reported. This release is then signaled by the emergence of a fluorescent signal. Importantly, the use of appropriate fluorophores has enabled not only this emerging fluorescence as a spatiotemporal marker for drug delivery but also has provided modalities useful in photodynamic, photothermal, and sonodynamic therapeutic applications. In this review we highlight recent work on theranostic fluorescent probes with a particular focus on probes that are activated in tumor microenvironments. We also summarize efforts to develop probes for other applications, such as neurodegenerative diseases and antibacterials. This review celebrates the diversity of designs reported to date, from discrete small-molecule systems to nanomaterials. Our aim is to provide insights into the potential clinical impact of this still-emerging research direction.
Collapse
Affiliation(s)
- Amit Sharma
- Amity
School of Chemical Sciences, Amity University
Punjab, Sector 82A, Mohali 140 306, India
| | - Peter Verwilst
- Rega
Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49, Box 1041, 3000 Leuven, Belgium
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Dandan Ma
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Nem Singh
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Jiyoung Yoo
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Yujin Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Ying Yang
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Jing-Hui Zhu
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Haiqiao Huang
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xi-Le Hu
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiao-Peng He
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- National
Center for Liver Cancer, the International Cooperation Laboratory
on Signal Transduction, Eastern Hepatobiliary
Surgery Hospital, Shanghai 200438, China
| | - Lintao Zeng
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- School
of Chemistry and Chemical Engineering, Henan
Normal University, Xinxiang 453007, China
| | - Xiaojun Peng
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- State
Key Laboratory of Fine Chemicals, Dalian
University of Technology, Dalian 116024, China
| | - Jonathan L. Sessler
- Department
of Chemistry, The University of Texas at
Austin, Texas 78712-1224, United
States
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- TheranoChem Incorporation, Seongbuk-gu, Seoul 02841, Korea
| |
Collapse
|
253
|
Chen H, Hou S, Zhang H, Zhou B, Xi H, Li X, Lufeng Z, Guo Q. MiR-375 impairs breast cancer cell stemness by targeting the KLF5/G6PD signaling axis. ENVIRONMENTAL TOXICOLOGY 2024. [PMID: 38470012 DOI: 10.1002/tox.24204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/13/2024]
Abstract
Recurrence of breast cancer may be due to the presence of breast cancer stem cells (BCSC). Abnormal tumor cell growth is closely associated with increased reactive oxygen species (ROS) and disruption of redox homeostasis, and BCSCs exhibit low levels of ROS. The detailed mechanism between the low levels of ROS in BCSCs and their maintenance of stemness characteristics has not been reported. A growing number of studies have shown that tumor development is often accompanied by metabolic reprogramming, which is an important hallmark of tumor cells. As the first rate-limiting enzyme of pentose phosphate pathway (PPP), the expression of G6PD is precisely regulated in tumor cells, and there is a certain correlation between PPP and BCSCs. MiR-375 has been shown to inhibit stem cell-like properties in breast cancer, but the exact mechanism is not clear. Here, KLF5, as a transcription factor, was identified to bind to the promoter of G6PD to promote its expression, whereas miR-375 inhibited the expression of KLF5 by binding to the 3'UTR region of KLF5 mRNA and thus reduced the expression of G6PD expression, inhibits PPP to reduce NADPH, and increases ROS levels in breast cancer cells, thereby weakening breast cancer cell stemness. Our study reveals the specific mechanism by which miR-375 targets the KLF5/G6PD signaling axis to diminish the stemness of breast cancer cells, providing a therapeutic strategy against BCSCs.
Collapse
Affiliation(s)
- Haitao Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shanshan Hou
- Department of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, People's Republic of China
| | - Hongwei Zhang
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Bing Zhou
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Huifang Xi
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Xiaofang Li
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Zheng Lufeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, People's Republic of China
| |
Collapse
|
254
|
Zhu Y, Zhao R, Feng L, Wang W, Xie Y, Ding H, Liu B, Dong S, Yang P, Lin J. Defect-Engineered Tin Disulfide Nanocarriers as "Precision-Guided Projectile" for Intensive Synergistic Therapy. SMALL METHODS 2024:e2400125. [PMID: 38461544 DOI: 10.1002/smtd.202400125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/25/2024] [Indexed: 03/12/2024]
Abstract
Nanoformulations with endogenous/exogenous stimulus-responsive characteristics show great potential in tumor cell elimination with minimal adverse effects and high precision. Herein, an intelligent nanotheranostic platform (denoted as TPZ@Cu-SnS2-x /PLL) for tumor microenvironment (TME) and near-infrared light (NIR) activated tumor-specific therapy is constructed. Copper (Cu) doping and the resulting sulfur vacancies can not only improve the response range of visible light but also improve the separation efficiency of photogenerated carriers and increase the carrier density, resulting in the ideal photothermal and photodynamic performance. Density functional theory calculations revealed that the introduction of Cu and resulting sulfur vacancies can induce electron redistribution, achieving favorable photogenerated electrons. After entering cells through endocytosis, the TPZ@Cu-SnS2-x /PLL nanocomposites show the pH responsivity property for the release of the TPZ selectively within the acidic TME, and the released Cu2+ can first interact with local glutathione (GSH) to deplete GSH with the production of Cu+ . Subsequently, the Cu+ -mediated Fenton-like reaction can decompose local hydrogen peroxide into hydroxyl radicals, which can also be promoted by hyperthermia derived from the photothermal effect for tumor cell apoptosis. The integration of photoacoustic/computed tomography imaging-guided NIR phototherapy, TPZ-induced chemotherapy, and GSH-elimination/hyperthermia enhanced chemodynamic therapy results in synergistic therapeutic outcomes without obvious systemic toxicity in vivo.
Collapse
Affiliation(s)
- Yanlin Zhu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ruoxi Zhao
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Lili Feng
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Wenzhuo Wang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ying Xie
- Key Laboratory of Functional Inorganic Material Chemistry, Ministry of Education, School of Chemistry and Materials Science, Heilongjiang University, Harbin, 150080, P. R. China
| | - He Ding
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Bin Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Shuming Dong
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|
255
|
Zhou Z, Li J, Ousmane D, Peng L, Yuan X, Wang J. Metabolic reprogramming directed by super-enhancers in tumors: An emerging landscape. Mol Ther 2024; 32:572-579. [PMID: 38327048 PMCID: PMC10928301 DOI: 10.1016/j.ymthe.2024.02.003] [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: 10/30/2023] [Revised: 01/09/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
Metabolic reprogramming is an essential hallmark of tumors, and metabolic abnormalities are strongly associated with the malignant phenotype of tumor cells. This is closely related to transcriptional dysregulation. Super-enhancers are extremely active cis-regulatory regions in the genome, and can amalgamate a complex set of transcriptional regulatory components that are crucial for establishing tumor cell identity, promoting tumorigenesis, and enhancing aggressiveness. In addition, alterations in metabolic signaling pathways are often accompanied by changes in super-enhancers. Presently, there is a surge in interest in the potential pathogenesis of various tumors through the transcriptional regulation of super-enhancers and oncogenic mutations in super-enhancers. In this review, we summarize the functions of super-enhancers, oncogenic signaling pathways, and tumor metabolic reprogramming. In particular, we focus on the role of the super-enhancer in tumor metabolism and its impact on metabolic reprogramming. This review also discusses the prospects and directions in the field of super-enhancer and metabolic reprogramming.
Collapse
Affiliation(s)
- Zongjiang Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jinghe Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
| | - Diabate Ousmane
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
| | - Li Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Xiaoqing Yuan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Junpu Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha, China; Ultrapathology (Biomedical Electron Microscopy) Center, Department of Pathology, Xiangya Hospital, Central South University, Changsha, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
256
|
Yan Y, Zhou S, Chen X, Yi Q, Feng S, Zhao Z, Liu Y, Liang Q, Xu Z, Li Z, Sun L. Suppression of ITPKB degradation by Trim25 confers TMZ resistance in glioblastoma through ROS homeostasis. Signal Transduct Target Ther 2024; 9:58. [PMID: 38438346 PMCID: PMC10912509 DOI: 10.1038/s41392-024-01763-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/05/2024] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
Temozolomide (TMZ) represents a standard-of-care chemotherapeutic agent in glioblastoma (GBM). However, the development of drug resistance constitutes a significant hurdle in the treatment of malignant glioma. Although specific innovative approaches, such as immunotherapy, have shown favorable clinical outcomes, the inherent invasiveness of most gliomas continues to make them challenging to treat. Consequently, there is an urgent need to identify effective therapeutic targets for gliomas to overcome chemoresistance and facilitate drug development. This investigation used mass spectrometry to examine the proteomic profiles of six pairs of GBM patients who underwent standard-of-care treatment and surgery for both primary and recurrent tumors. A total of 648 proteins exhibiting significant differential expression were identified. Gene Set Enrichment Analysis (GSEA) unveiled notable alterations in pathways related to METABOLISM_OF_LIPIDS and BIOLOGICAL_OXIDATIONS between the primary and recurrent groups. Validation through glioma tissue arrays and the Xiangya cohort confirmed substantial upregulation of inositol 1,4,5-triphosphate (IP3) kinase B (ITPKB) in the recurrence group, correlating with poor survival in glioma patients. In TMZ-resistant cells, the depletion of ITPKB led to an increase in reactive oxygen species (ROS) related to NADPH oxidase (NOX) activity and restored cell sensitivity to TMZ. Mechanistically, the decreased phosphorylation of the E3 ligase Trim25 at the S100 position in recurrent GBM samples accounted for the weakened ITPKB ubiquitination. This, in turn, elevated ITPKB stability and impaired ROS production. Furthermore, ITPKB depletion or the ITPKB inhibitor GNF362 effectively overcome TMZ chemoresistance in a glioma xenograft mouse model. These findings reveal a novel mechanism underlying TMZ resistance and propose ITPKB as a promising therapeutic target for TMZ-resistant GBM.
Collapse
Affiliation(s)
- Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shangjun Zhou
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiaoli Yi
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Songshan Feng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zijin Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Zhi Li
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China.
- Institute of Cancer Research, National Clinical Research Center for Geriatric Disorders (Xiangya), Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Lunquan Sun
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China.
- Institute of Cancer Research, National Clinical Research Center for Geriatric Disorders (Xiangya), Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
257
|
Tang Q, Li X, Sun CR. Predictive value of serum albumin levels on cancer survival: a prospective cohort study. Front Oncol 2024; 14:1323192. [PMID: 38500655 PMCID: PMC10944876 DOI: 10.3389/fonc.2024.1323192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/22/2024] [Indexed: 03/20/2024] Open
Abstract
Background Serum albumin levels and cancer mortality are closely related, yet large-sample studies encompassing a broad spectrum of cancer types are lacking. Methods This study encompassed patients diagnosed with cancer across the continuous 10 cycles of NHANES surveys from 1999 to 2018. The study population was stratified into two groups based on median albumin levels (≤ 4.2g/dL and > 4.2 g/dL) or cancer aggressiveness (well-survived cancers and poorly-survived cancers). Survival rates were estimated using the Kaplan-Meier method. The Cox proportional hazards model was employed to evaluate the association between serum albumin levels and cancer mortality. Restricted cubic spline (RCS) analysis was conducted to assess the nonlinear relationship between serum albumin levels and the risk of cancer mortality. Results Kaplan-Meier curves demonstrated that patients with albumin levels ≤ 4.2 g/dL exhibited lower survival rates compared to those with levels > 4.2 g/dL, irrespective of cancer aggressiveness. Following adjustment for confounders, decreased albumin levels were associated with an elevated risk of cancer mortality across all groups [all cancers, HR (95%CI) = 2.03(1.73, 2.37); well survived cancers, HR (95%CI) = 1.78(1.38, 2.32); and poorly survived cancers, HR (95%CI) = 1.99(1.64, 2.42)]. RCS analyses revealed a stable nonlinear negative association between albumin levels and cancer mortality in all groups, regardless of confounder adjustment. Conclusion Low serum albumin levels predict higher cancer mortality. Furthermore, a nonlinear negative association was observed between serum albumin levels and the risk of cancer mortality.
Collapse
Affiliation(s)
- Quan Tang
- Department of General Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Xu Li
- Division of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chun-Rong Sun
- Department of General Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
258
|
Wu X, Li Y, Wen M, Xie Y, Zeng K, Liu YN, Chen W, Zhao Y. Nanocatalysts for modulating antitumor immunity: fabrication, mechanisms and applications. Chem Soc Rev 2024; 53:2643-2692. [PMID: 38314836 DOI: 10.1039/d3cs00673e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Immunotherapy harnesses the inherent immune system in the body to generate systemic antitumor immunity, offering a promising modality for defending against cancer. However, tumor immunosuppression and evasion seriously restrict the immune response rates in clinical settings. Catalytic nanomedicines can transform tumoral substances/metabolites into therapeutic products in situ, offering unique advantages in antitumor immunotherapy. Through catalytic reactions, both tumor eradication and immune regulation can be simultaneously achieved, favoring the development of systemic antitumor immunity. In recent years, with advancements in catalytic chemistry and nanotechnology, catalytic nanomedicines based on nanozymes, photocatalysts, sonocatalysts, Fenton catalysts, electrocatalysts, piezocatalysts, thermocatalysts and radiocatalysts have been rapidly developed with vast applications in cancer immunotherapy. This review provides an introduction to the fabrication of catalytic nanomedicines with an emphasis on their structures and engineering strategies. Furthermore, the catalytic substrates and state-of-the-art applications of nanocatalysts in cancer immunotherapy have also been outlined and discussed. The relationships between nanostructures and immune regulating performance of catalytic nanomedicines are highlighted to provide a deep understanding of their working mechanisms in the tumor microenvironment. Finally, the challenges and development trends are revealed, aiming to provide new insights for the future development of nanocatalysts in catalytic immunotherapy.
Collapse
Affiliation(s)
- Xianbo Wu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Yuqing Li
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Mei Wen
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Yongting Xie
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Ke Zeng
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - You-Nian Liu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Wansong Chen
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore.
| |
Collapse
|
259
|
Wang H, Liu J, Zhang Z, Peng J, Wang Z, Yang L, Wang X, Hu S, Hong L. β-Sitosterol targets ASS1 for Nrf2 ubiquitin-dependent degradation, inducing ROS-mediated apoptosis via the PTEN/PI3K/AKT signaling pathway in ovarian cancer. Free Radic Biol Med 2024; 214:137-157. [PMID: 38364944 DOI: 10.1016/j.freeradbiomed.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 02/18/2024]
Abstract
The exploration of drugs derived from natural sources holds significant promise in addressing current limitations in ovarian cancer (OC) treatments. While previous studies have highlighted the remarkable anti-cancer properties of the natural compound β-sitosterol (SIT) across various tumors, its specific role in OC treatment remains unexplored. This study aims to investigate the anti-tumor activity of SIT in OC using in vitro and in vivo models, delineate potential mechanisms, and establish a preclinical theoretical foundation for future clinical trials, thus fostering further research. Utilizing network pharmacology, we pinpoint SIT as a promising candidate for OC treatment and predict its potential targets and pathways. Through a series of in vitro and in vivo experiments, we unveil a novel mechanism through which SIT mitigates the malignant biological behaviors of OC cells by modulating redox status. Specifically, SIT selectively targets argininosuccinate synthetase 1 (ASS1), a protein markedly overexpressed in OC tissues and cells. Inhibiting ASS1, SIT enhances the interaction between Nrf2 and Keap1, instigating the ubiquitin-dependent degradation of Nrf2, subsequently diminishing the transcriptional activation of downstream antioxidant genes HO-1 and NQO1. The interruption of the antioxidant program by SIT results in the substantial accumulation of reactive oxygen species (ROS) in OC cells. This, in turn, upregulates PTEN, exerting negative regulation on the phosphorylation activation of AKT. The suppression of AKT signaling disrupted downstream pathways associated with cell cycle, cell survival, apoptosis, migration, and invasion, ultimately culminating in the death of OC cells. Our research uncovers new targets and mechanisms of SIT against OC, contributing to the existing knowledge on the anti-tumor effects of natural products in the context of OC. Additionally, this research unveils a novel role of ASS1 in regulating the Nrf2-mediated antioxidant program and governing redox homeostasis in OC, providing a deeper understanding of this complex disease.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Jingchun Liu
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Zihui Zhang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Jiaxin Peng
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Zhi Wang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Lian Yang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Xinqi Wang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Siyuan Hu
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Li Hong
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| |
Collapse
|
260
|
Wu K, El Zowalaty AE, Sayin VI, Papagiannakopoulos T. The pleiotropic functions of reactive oxygen species in cancer. NATURE CANCER 2024; 5:384-399. [PMID: 38531982 DOI: 10.1038/s43018-024-00738-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 01/19/2024] [Indexed: 03/28/2024]
Abstract
Cellular redox homeostasis is an essential, dynamic process that ensures the balance between reducing and oxidizing reactions within cells and thus has implications across all areas of biology. Changes in levels of reactive oxygen species can disrupt redox homeostasis, leading to oxidative or reductive stress that contributes to the pathogenesis of many malignancies, including cancer. From transformation and tumor initiation to metastatic dissemination, increasing reactive oxygen species in cancer cells can paradoxically promote or suppress the tumorigenic process, depending on the extent of redox stress, its spatiotemporal characteristics and the tumor microenvironment. Here we review how redox regulation influences tumorigenesis, highlighting therapeutic opportunities enabled by redox-related alterations in cancer cells.
Collapse
Affiliation(s)
- Katherine Wu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Ahmed Ezat El Zowalaty
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Volkan I Sayin
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA.
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
261
|
Wang B, Lu ZN, Song MX, He XW, Hu ZC, Liang HF, Lu HW, Chen Q, Liang B, Yi T, Wei P, Jiang LB, Dong J. Single-Component Dual-Functional Autoboost Strategy by Dual Photodynamic and Cyclooxygenase-2 Inhibition for Lung Cancer and Spinal Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303981. [PMID: 38224203 DOI: 10.1002/advs.202303981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 12/24/2023] [Indexed: 01/16/2024]
Abstract
Coloading adjuvant drugs or biomacromolecules with photosensitizers into nanoparticles to enhance the efficiency of photodynamic therapy (PDT) is a common strategy. However, it is difficult to load positively charged photosensitizers and negatively charged adjuvants into the same nanomaterial and further regulate drug release simultaneously. Herein, a single-component dual-functional prodrug strategy is reported for tumor treatment specifically activated by tumor microenvironment (TME)-generated HOCl. A representative prodrug (DHU-CBA2) is constructed using indomethacin grafted with methylene blue (MB). DHU-CBA2 exhibited high sensitivity toward HOCl and achieved simultaneous release of dual drugs in vitro and in vivo. DHU-CBA2 shows effective antitumor activity against lung cancer and spinal metastases via PDT and cyclooxygenase-2 (COX-2) inhibition. Mechanistically, PDT induces immunogenic cell death but stimulates the gene encoding COX-2. Downstream prostaglandins E2 and Indoleamine 2,3 dioxygenase 1 (IDO1) mediate immune escape in the TME, which is rescued by the simultaneous release of indomethacin. DHU-CBA2 promotes infiltration and function of CD8+ T cells, thus inducing a robust antitumor immune response. This work provides an autoboost strategy for a single-component dual-functional prodrug activated by TME-specific HOCl, thereby achieving favorable tumor treatment via the synergistic therapy of PDT and a COX-2 inhibitor.
Collapse
Affiliation(s)
- Ben Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhen-Ni Lu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Meng-Xiong Song
- Department of Orthopedics Surgery, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Xiao-Wen He
- Department of Orthopaedic Surgery, Shanghai Baoshan District Wusong Center Hospital, Fudan University, Shanghai, 200940, China
| | - Zhi-Chao Hu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hai-Feng Liang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hong-Wei Lu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qing Chen
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Bing Liang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tao Yi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Peng Wei
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Li-Bo Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Orthopaedic Surgery, Shanghai Baoshan District Wusong Center Hospital, Fudan University, Shanghai, 200940, China
| |
Collapse
|
262
|
Fei J, Liu Y, Zeng Y, Yang M, Chen S, Duan X, Lu L, Chen M. Cancer diagnosis and treatment platform based on manganese-based nanomaterials. Front Bioeng Biotechnol 2024; 12:1363569. [PMID: 38497051 PMCID: PMC10940866 DOI: 10.3389/fbioe.2024.1363569] [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: 12/31/2023] [Accepted: 01/29/2024] [Indexed: 03/19/2024] Open
Abstract
Cancer is a leading cause of death worldwide, and the development of new diagnostic and treatment methods is crucial. Manganese-based nanomaterials (MnNMs) have emerged as a focal point in the field of cancer diagnosis and treatment due to their multifunctional properties. These nanomaterials have been extensively explored as contrast agents for various imaging technologies such as magnetic resonance imaging (MRI), photoacoustic imaging (PAI), and near-infrared fluorescence imaging (NIR-FL). The use of these nanomaterials has significantly enhanced the contrast for precise tumor detection and localization. Moreover, MnNMs have shown responsiveness to the tumor microenvironment (TME), enabling innovative approaches to cancer treatment. This review provides an overview of the latest developments of MnNMs and their potential applications in tumor diagnosis and therapy. Finally, potential challenges and prospects of MnNMs in clinical applications are discussed. We believe that this review would serve as a valuable resource for guiding further research on the application of manganese nanomaterials in cancer diagnosis and treatment, addressing the current limitations, and proposing future research directions.
Collapse
Affiliation(s)
- Jia Fei
- Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, China
| | - Yanyan Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong, China
| | - Ya Zeng
- Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, China
| | - Mingqi Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong, China
| | - Shanshan Chen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong, China
| | - Xiaobing Duan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong, China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong, China
| | - Muhe Chen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong, China
| |
Collapse
|
263
|
Wang Y, Jia X, An S, Yin W, Huang J, Jiang X. Nanozyme-Based Regulation of Cellular Metabolism and Their Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2301810. [PMID: 37017586 DOI: 10.1002/adma.202301810] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Metabolism is the sum of the enzyme-dependent chemical reactions, which produces energy in catabolic process and synthesizes biomass in anabolic process, exhibiting high similarity in mammalian cell, microbial cell, and plant cell. Consequently, the loss or gain of metabolic enzyme activity greatly affects cellular metabolism. Nanozymes, as emerging enzyme mimics with diverse functions and adjustable catalytic activities, have shown attractive potential for metabolic regulation. Although the basic metabolic tasks are highly similar for the cells from different species, the concrete metabolic pathway varies with the intracellular structure of different species. Here, the basic metabolism in living organisms is described and the similarities and differences in the metabolic pathways among mammalian, microbial, and plant cells and the regulation mechanism are discussed. The recent progress on regulation of cellular metabolism mainly including nutrient uptake and utilization, energy production, and the accompanied redox reactions by different kinds of oxidoreductases and their applications in the field of disease therapy, antimicrobial therapy, and sustainable agriculture is systematically reviewed. Furthermore, the prospects and challenges of nanozymes in regulating cell metabolism are also discussed, which broaden their application scenarios.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Xiaodan Jia
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Shangjie An
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Wenbo Yin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Jiahao Huang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Xiue Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
264
|
Xu F, Li M, Qian Q, Chen L, Yang Y, Ji TF, Li JG. β-acetoxyisovalerylalkannin suppresses proliferation and induces ROS-based mitochondria-mediated apoptosis in human melanoma cells. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:372-386. [PMID: 37310856 DOI: 10.1080/10286020.2023.2221648] [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: 04/23/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023]
Abstract
β-acetoxyisovalerylalkannin (β-AIVA) is one of shikonin/alkannin derivative, which were mainly extracted from Boraginaceae family. The effects of β-AIVA on human melanoma A375 cells and U918 cells were investigated in vitro. The CCK-8 assay showed that β-AIVA inhibited proliferation of cells. Results from flow cytometry, ROS assay and JC-1 assay showed that β-AIVA increased late apoptosis rate, induced the production of ROS and promoted mitochondrial depolarization in cells. β-AIVA regulated expressions of BAX and Bcl-2 proteins, and increased the expression of cleaved caspase-9 and cleaved caspase-3. These findings suggest that β-AIVA may be a potential therapeutic drug for treating melanoma.
Collapse
Affiliation(s)
- Fang Xu
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Min Li
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Qian Qian
- Department of Pharmacy, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Ling Chen
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Ying Yang
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Teng-Fei Ji
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jian-Guang Li
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
- Xinjiang University of Science & Technology, Korla 841899, China
| |
Collapse
|
265
|
Zhang Z, Zhao Q, Wang Z, Xu F, Liu Y, Guo Y, Li C, Liu T, Zhao Y, Tang X, Zhang J. Hepatocellular carcinoma cells downregulate NADH:Ubiquinone Oxidoreductase Subunit B3 to maintain reactive oxygen species homeostasis. Hepatol Commun 2024; 8:e0395. [PMID: 38437062 PMCID: PMC10914236 DOI: 10.1097/hc9.0000000000000395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/02/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND HCC is a leading cause of cancer-related death. The role of reactive oxygen species (ROS) in HCC remains elusive. Since a primary ROS source is the mitochondrial electron transport chain complex Ι and the NADH:ubiquinone Oxidoreductase Subunit B3 (NDUFB3), a complex I subunit, is critical for complex I assembly and regulates the associated ROS production, we hypothesize that some HCCs progress by hijacking NDUFB3 to maintain ROS homeostasis. METHODS NDUFB3 in human HCC lines was either knocked down or overexpressed. The cells were then analyzed in vitro for proliferation, migration, invasiveness, colony formation, complex I activity, ROS production, oxygen consumption, apoptosis, and cell cycle. In addition, the in vivo growth of the cells was evaluated in nude mice. Moreover, the role of ROS in the NDUFB3-mediated changes in the HCC lines was determined using cellular and mitochondrion-targeted ROS scavengers. RESULTS HCC tissues showed reduced NDUFB3 protein expression compared to adjacent healthy tissues. In addition, NDUFB3 knockdown promoted, while its overexpression suppressed, HCC cells' growth, migration, and invasiveness. Moreover, NDUFB3 knockdown significantly decreased, whereas its overexpression increased complex I activity. Further studies revealed that NDUFB3 overexpression elevated mitochondrial ROS production, causing cell apoptosis, as manifested by the enhanced expressions of proapoptotic molecules and the suppressed expression of the antiapoptotic molecule B cell lymphoma 2. Finally, our data demonstrated that the apoptosis was due to the activation of the c-Jun N-terminal kinase (JNK) signaling pathway and cell cycle arrest at G0/G1 phase. CONCLUSIONS Because ROS plays essential roles in many biological processes, such as aging and cancers, our findings suggest that NDFUB3 can be targeted for treating HCC and other human diseases.
Collapse
Affiliation(s)
- Zhendong Zhang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- BGI College, Zhengzhou University, Zhengzhou, China
| | - Qianwei Zhao
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Zexuan Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- BGI College, Zhengzhou University, Zhengzhou, China
| | - Fang Xu
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Yixian Liu
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Yaoyu Guo
- BGI College, Zhengzhou University, Zhengzhou, China
| | - Chenglong Li
- School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ting Liu
- BGI College, Zhengzhou University, Zhengzhou, China
| | - Ying Zhao
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaolei Tang
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, New York, USA
- Department of Medicine, Division of Regenerative Medicine, School of Medicine, Loma Linda University, Loma Linda, California, USA
- Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Jintao Zhang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
266
|
Shiraishi Y, Matsuya Y, Fukunaga H. Possible mechanisms and simulation modeling of FLASH radiotherapy. Radiol Phys Technol 2024; 17:11-23. [PMID: 38184508 DOI: 10.1007/s12194-023-00770-x] [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: 05/23/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 01/08/2024]
Abstract
FLASH radiotherapy (FLASH-RT) has great potential to improve patient outcomes. It delivers radiation doses at an ultra-high dose rate (UHDR: ≥ 40 Gy/s) in a single instant or a few pulses. Much higher irradiation doses can be administered to tumors with FLASH-RT than with conventional dose rate (0.01-0.40 Gy/s) radiotherapy. UHDR irradiation can suppress toxicity in normal tissues while sustaining antitumor efficiency, which is referred to as the FLASH effect. However, the mechanisms underlying the effects of the FLASH remain unclear. To clarify these mechanisms, the development of simulation models that can contribute to treatment planning for FLASH-RT is still underway. Previous studies indicated that transient oxygen depletion or augmented reactions between secondary reactive species produced by irradiation may be involved in this process. To discuss the possible mechanisms of the FLASH effect and its clinical potential, we summarized the physicochemical, chemical, and biological perspectives as well as the development of simulation modeling for FLASH-RT.
Collapse
Affiliation(s)
- Yuta Shiraishi
- Graduate School of Health Sciences, Hokkaido University, N12 W5 Kita-Ku, Sapporo, Hokkaido, 060-0812, Japan
- Faculty of Health Sciences, Japan Healthcare University, 3-11-1-50 Tsukisamu-Higashi, Toyohira-Ku, Sapporo, Hokkaido, 062-0053, Japan
| | - Yusuke Matsuya
- Faculty of Health Sciences, Hokkaido University, N12 W5 Kita-Ku, Sapporo, Hokkaido, 060-0812, Japan
| | - Hisanori Fukunaga
- Faculty of Health Sciences, Hokkaido University, N12 W5 Kita-Ku, Sapporo, Hokkaido, 060-0812, Japan.
| |
Collapse
|
267
|
He Z, Du J, Wang Q, Chen G, Li X, Zhang Z, Wang S, Jing W, Miao Q, Li Y, Miao Y, Wu J. Dye-augmented bandgap engineering of a degradable cascade nanoreactor for tumor immune microenvironment-enhanced dynamic phototherapy of breast cancer. Acta Biomater 2024; 176:390-404. [PMID: 38244657 DOI: 10.1016/j.actbio.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/31/2023] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
Non-invasive precision tumor dynamic phototherapy has broad application prospects. Traditional semiconductor materials have low photocatalytic activity and low reactive oxygen species (ROS) production rate due to their wide band gap, resulting in unsatisfactory phototherapy efficacy for tumor treatment. Employing the dye-sensitization mechanism can significantly enhance the catalytic activity of the materials. We develop a multifunctional nanoplatform (BZP) by leveraging the benefits of bismuth-based semiconductor nanomaterials. BZP possesses robust ROS generation and remarkable near-infrared photothermal conversion capabilities for improving tumor immune microenvironment and achieving superior phototherapy sensitization. BZP produces highly cytotoxic ROS species via the photocatalytic process and cascade reaction, amplifying the photocatalytic therapy effect. Moreover, the simultaneous photothermal effect during the photocatalytic process facilitates the improvement of therapeutic efficacy. Additionally, BZP-mediated phototherapy can trigger the programmed death of tumor cells, stimulate dendritic cell maturation and T cell activation, modulate the tumor immune microenvironment, and augment the therapeutic effect. Hence, this study demonstrates a promising research paradigm for tumor immune microenvironment-improved phototherapy. STATEMENT OF SIGNIFICANCE: Through the utilization of dye sensitization and rare earth doping techniques, we have successfully developed a biodegradable bismuth-based semiconductor nanocatalyst (BZP). Upon optical excitation, the near-infrared dye incorporated within BZP promptly generates free electrons, which, under the influence of the Fermi energy level, undergo transfer to BiF3 within BZP, thereby facilitating the effective separation of electron-hole pairs and augmenting the catalytic capability for reactive oxygen species (ROS) generation. Furthermore, a cascade reaction mechanism generates highly cytotoxic ROS, which synergistically depletes intracellular glutathione, thereby intensifying oxidative stress. Ultimately, this dual activation strategy, combining oxidative and thermal damage, holds significant potential for tumor immunotherapy.
Collapse
Affiliation(s)
- Zongyan He
- Department of Anesthesiology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Bismuth Science, School of Materials and Chemistry, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jun Du
- Institute of Bismuth Science, School of Materials and Chemistry, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Qian Wang
- Institute of Bismuth Science, School of Materials and Chemistry, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Guobo Chen
- Institute of Bismuth Science, School of Materials and Chemistry, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Xueyu Li
- Institute of Bismuth Science, School of Materials and Chemistry, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Zheng Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Shanhou Wang
- Institute of Bismuth Science, School of Materials and Chemistry, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Wenxuan Jing
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Qing Miao
- Department of Anesthesiology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Yuhao Li
- Institute of Bismuth Science, School of Materials and Chemistry, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yuqing Miao
- Institute of Bismuth Science, School of Materials and Chemistry, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jingxiang Wu
- Department of Anesthesiology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
268
|
Wang B, Fei X, Yin HF, Xu XN, Zhu JJ, Guo ZY, Wu JW, Zhu XS, Zhang Y, Xu Y, Yang Y, Chen LS. Photothermal-Controllable Microneedles with Antitumor, Antioxidant, Angiogenic, and Chondrogenic Activities to Sequential Eliminate Tracheal Neoplasm and Reconstruct Tracheal Cartilage. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309454. [PMID: 38098368 DOI: 10.1002/smll.202309454] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Indexed: 03/16/2024]
Abstract
The optimal treatment for tracheal tumors necessitates sequential tumor elimination and tracheal cartilage reconstruction. This study introduces an innovative inorganic nanosheet, MnO2 /PDA@Cu, comprising manganese dioxide (MnO2 ) loaded with copper ions (Cu) through in situ polymerization using polydopamine (PDA) as an intermediary. Additionally, a specialized methacrylic anhydride modified decellularized cartilage matrix (MDC) hydrogel with chondrogenic effects is developed by modifying a decellularized cartilage matrix with methacrylic anhydride. The MnO2 /PDA@Cu nanosheet is encapsulated within MDC-derived microneedles, creating a photothermal-controllable MnO2 /PDA@Cu-MDC microneedle. Effectiveness evaluation involved deep insertion of the MnO2 /PDA@Cu-MDC microneedle into tracheal orthotopic tumor in a murine model. Under 808 nm near-infrared irradiation, facilitated by PDA, the microneedle exhibited rapid overheating, efficiently eliminating tumors. PDA's photothermal effects triggered controlled MnO2 and Cu release. The MnO2 nanosheet acted as a potent inorganic nanoenzyme, scavenging reactive oxygen species for an antioxidant effect, while Cu facilitated angiogenesis. This intervention enhanced blood supply at the tumor excision site, promoting stem cell enrichment and nutrient provision. The MDC hydrogel played a pivotal role in creating a chondrogenic niche, fostering stem cells to secrete cartilaginous matrix. In conclusion, the MnO2 /PDA@Cu-MDC microneedle is a versatile platform with photothermal control, sequentially combining antitumor, antioxidant, pro-angiogenic, and chondrogenic activities to orchestrate precise tracheal tumor eradication and cartilage regeneration.
Collapse
Affiliation(s)
- B Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - X Fei
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - H F Yin
- Department of Infection Management, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - X N Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - J J Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Z Y Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - J W Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - X S Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Y Zhang
- Department of Orthopedics, Shanghai Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China
| | - Y Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Y Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
- Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - L S Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| |
Collapse
|
269
|
Ren X, Shi P, Su J, Wei T, Li J, Hu Y, Wu C. Loss of Myo19 increases metastasis by enhancing microenvironmental ROS gradient and chemotaxis. EMBO Rep 2024; 25:971-990. [PMID: 38279020 PMCID: PMC10933354 DOI: 10.1038/s44319-023-00052-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 12/10/2023] [Accepted: 12/19/2023] [Indexed: 01/28/2024] Open
Abstract
Tumor metastasis involves cells migrating directionally in response to external chemical signals. Reactive oxygen species (ROS) in the form of H2O2 has been demonstrated as a chemoattractant for neutrophils but its spatial characteristics in tumor microenvironment and potential role in tumor cell dissemination remain unknown. Here we investigate the spatial ROS distribution in 3D tumor spheroids and identify a ROS concentration gradient in spheroid periphery, which projects into a H2O2 gradient in tumor microenvironment. We further reveal the role of H2O2 gradient to induce chemotaxis of tumor cells by activating Src and subsequently inhibiting RhoA. Finally, we observe that the absence of mitochondria cristae remodeling proteins including the mitochondria-localized actin motor Myosin 19 (Myo19) enhances ROS gradient and promotes tumor dissemination. Myo19 downregulation is seen in many tumors, and Myo19 expression is negatively associated with tumor metastasis in vivo. Together, our study reveals the chemoattractant role of tumor microenvironmental ROS and implies the potential impact of mitochondria cristae disorganization on tumor invasion and metastasis.
Collapse
Affiliation(s)
- Xiaoyu Ren
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China
| | - Peng Shi
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China.
- International Cancer Institute, Peking University, Beijing, 100191, China.
| | - Jing Su
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Tonghua Wei
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China
| | - Jiayi Li
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China
| | - Yiping Hu
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China
| | - Congying Wu
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China.
- International Cancer Institute, Peking University, Beijing, 100191, China.
| |
Collapse
|
270
|
Liu Z, Liu X, Zhang W, Gao R, Wei H, Yu CY. Current advances in modulating tumor hypoxia for enhanced therapeutic efficacy. Acta Biomater 2024; 176:1-27. [PMID: 38232912 DOI: 10.1016/j.actbio.2024.01.010] [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: 08/07/2023] [Revised: 12/08/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Hypoxia is a common feature of most solid tumors, which promotes the proliferation, invasion, metastasis, and therapeutic resistance of tumors. Researchers have been developing advanced strategies and nanoplatforms to modulate tumor hypoxia to enhance therapeutic effects. A timely review of this rapidly developing research topic is therefore highly desirable. For this purpose, this review first introduces the impact of hypoxia on tumor development and therapeutic resistance in detail. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are also systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We provide a detailed discussion of the rationale and research progress of these strategies. Through a review of current trends, it is hoped that this comprehensive overview can provide new prospects for clinical application in tumor treatment. STATEMENT OF SIGNIFICANCE: As a common feature of most solid tumors, hypoxia significantly promotes tumor progression. Advanced nanoplatforms have been developed to modulate tumor hypoxia to enhanced therapeutic effects. In this review, we first introduce the impact of hypoxia on tumor progression. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We discuss the rationale and research progress of the above strategies in detail, and finally introduce future challenges for treatment of hypoxic tumors. By reviewing the current trends, this comprehensive overview can provide new prospects for clinical translatable tumor therapy.
Collapse
Affiliation(s)
- Zihan Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xinping Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Wei Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ruijie Gao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
271
|
Feng Y, An Q, Zhao Z, Wu M, Yang C, Liang W, Xu X, Jiang T, Zhang G. Beta-elemene: A phytochemical with promise as a drug candidate for tumor therapy and adjuvant tumor therapy. Biomed Pharmacother 2024; 172:116266. [PMID: 38350368 DOI: 10.1016/j.biopha.2024.116266] [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: 11/22/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND β-Elemene (IUPAC name: (1 S,2 S,4 R)-1-ethenyl-1-methyl-2,4-bis(prop-1-en-2-yl) cyclohexane), is a natural compound found in turmeric root. Studies have demonstrated its diverse biological functions, including its anti-tumor properties, which have been extensively investigated. However, these have not yet been reviewed. The aim of this review was to provide a comprehensive summary of β-elemene research, with respect to disease treatment. METHODS β-Elemene-related articles were found in PubMed, ScienceDirect, and Google Scholar databases to systematically summarize its structure, pharmacokinetics, metabolism, and pharmacological activity. We also searched the Traditional Chinese Medicine System Pharmacology database for therapeutic targets of β-elemene. We further combined these targets with the relevant literature for KEGG and GO analyses. RESULTS Studies on the molecular mechanisms underlying β-elemene activity indicate that it regulates multiple pathways, including STAT3, MAPKs, Cyclin-dependent kinase 1/cyclin B, Notch, PI3K/AKT, reactive oxygen species, METTL3, PTEN, p53, FAK, MMP, TGF-β/Smad signaling. Through these molecular pathways, β-elemene has been implicated in tumor cell proliferation, apoptosis, migration, and invasion and improving the immune microenvironment. Additionally, β-elemene increases chemotherapeutic drug sensitivity and reverses resistance by inhibiting DNA damage repair and regulating pathways including CTR1, pak1, ERK1/2, ABC transporter protein, Prx-1 and ERCC-1. Nonetheless, owing to its lipophilicity and low bioavailability, additional structural modifications could improve the efficacy of this drug. CONCLUSION β-Elemene exhibits low toxicity with good safety, inhibiting various tumor types via diverse mechanisms in vivo and in vitro. When combined with chemotherapeutic drugs, it enhances efficacy, reduces toxicity, and improves tumor killing. Thus, β-elemene has vast potential for research and development.
Collapse
Affiliation(s)
- Yewen Feng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Qingwen An
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Zhengqi Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Mengting Wu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Chuqi Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - WeiYu Liang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Xuefei Xu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Tao Jiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China.
| | - Guangji Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China.
| |
Collapse
|
272
|
Dong Q, Turdu G, Akber Aisa H, Yili A. Arenobufagin, isolated from Bufo viridis toad venom, inhibits A549 cells proliferation by inducing apoptosis and G2/M cell cycle arrest. Toxicon 2024; 240:107641. [PMID: 38331108 DOI: 10.1016/j.toxicon.2024.107641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/11/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Lung cancer is a significant contributor to cancer morbidity and mortality globally. Arenobufagin, a compound derived from Bufo viridis toad venom, has demonstrated the ability to inhibit cell growth in various cancer cell lines. However, our understanding of the role and mechanism of arenobufagin in lung cancer remains incomplete, necessitating further researches to fully elucidate its action mechanism. In this study, we further explored the impact of arenobufagin on A549 cells. The results revealed that it exerted a potent cytotoxic effect on A549 cells by inhibiting cell colony formation, promoting cell apoptosis, increasing reactive oxygen species (ROS) levels, and arresting A549 cells in G2/M phase. Collectively, our findings suggested that arenobufagin may have potential as a future therapeutic for lung cancer treatment.
Collapse
Affiliation(s)
- Qiang Dong
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, and the Key Laboratory of Chemistry of Plant Resources in Arid Regions Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, South Beijing Road 40-1, Urumqi, 830011, People's Republic of China; University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Gulmira Turdu
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, and the Key Laboratory of Chemistry of Plant Resources in Arid Regions Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, South Beijing Road 40-1, Urumqi, 830011, People's Republic of China; University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Haji Akber Aisa
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, and the Key Laboratory of Chemistry of Plant Resources in Arid Regions Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, South Beijing Road 40-1, Urumqi, 830011, People's Republic of China
| | - Abulimiti Yili
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, and the Key Laboratory of Chemistry of Plant Resources in Arid Regions Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, South Beijing Road 40-1, Urumqi, 830011, People's Republic of China.
| |
Collapse
|
273
|
Lou J, Jin M, Zhou C, Fan Y, Ni L, Mao Y, Shen H, Li J, Zhang H, Fu C, Mao X, Chen Y, Zhong J, Zhou K, Wang L, Wu J. Ezrin inhibition alleviates oxidative stress and pyroptosis via regulating TRPML1-calcineurin axis mediated enhancement of autophagy in spinal cord injury. Free Radic Biol Med 2024; 212:133-148. [PMID: 38142951 DOI: 10.1016/j.freeradbiomed.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Spinal cord injury (SCI) presents profound ramifications for patients, leading to diminished motor and sensory capabilities distal to the lesion site. Once SCI occurs, it not only causes great physical and psychological problems for patients but also imposes a heavy economic burden. Ezrin is involved in various cellular processes, including signal transduction, cell death, inflammation, chemotherapy resistance and the stress response. However, whether Ezrin regulates functional repair after SCI and its underlying mechanism has not been elucidated. Here, our results showed that there is a marked augmentation of Ezrin levels within neurons and Ezrin inhibition markedly diminished glial scarring and bolstered functional recuperation after SCI. RNA sequencing indicated the potential involvement of pyroptosis, oxidative stress and autophagy in the enhancement of functional recovery upon reduced Ezrin expression. Moreover, the inhibition of Ezrin expression curtailed pyroptosis and oxidative stress by amplifying autophagy. Our studies further demonstrated that Ezrin inhibition promoted autophagy by increasing TFEB activity via the Akt-TRPML1-calcineurin pathway. Finally, we concluded that inhibiting Ezrin expression alleviates pyroptosis and oxidative stress by enhancing TFEB-driven autophagy, thereby promoting functional recovery after SCI, which may be a promising therapeutic target for SCI treatment.
Collapse
Affiliation(s)
- Junsheng Lou
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Mengran Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Conghui Zhou
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Yunpeng Fan
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Libin Ni
- Department of Orthopaedics, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, No. 1229 Gudun Road, Hangzhou, 310030, Zhejiang, China
| | - Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Honghao Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Jiafeng Li
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Haojie Zhang
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Chunyan Fu
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Xingjia Mao
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yingying Chen
- Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinjie Zhong
- Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| | - Linlin Wang
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Tarim University, School of Medicine, Alaer, 843300, China.
| | - Junsong Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
274
|
Sahu P, Camarillo IG, Sundararajan R. Efficacy of metformin and electrical pulses in breast cancer MDA-MB-231 cells. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:54-73. [PMID: 38464382 PMCID: PMC10918234 DOI: 10.37349/etat.2024.00204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/30/2023] [Indexed: 03/12/2024] Open
Abstract
Aim Triple-negative breast cancer (TNBC) is a very aggressive subset of breast cancer, with limited treatment options, due to the lack of three commonly targeted receptors, which merits the need for novel treatments for TNBC. Towards this need, the use of metformin (Met), the most widely used type-2 diabetes drug worldwide, was explored as a repurposed anticancer agent. Cancer being a metabolic disease, the modulation of two crucial metabolites, glucose, and reactive oxygen species (ROS), is studied in MDA-MB-231 TNBC cells, using Met in the presence of electrical pulses (EP) to enhance the drug efficacy. Methods MDA-MB-231, human TNBC cells were treated with Met in the presence of EP, with various concentrations Met of 1 mmol/L, 2.5 mmol/L, 5 mmol/L, and 10 mmol/L. EP of 500 V/cm, 800 V/cm, and 1,000 V/cm (with a pulse width of 100 µs at 1 s intervals) were applied to TNBC and the impact of these two treatments was studied. Various assays, including cell viability, microscopic inspection, glucose, ROS, and wound healing assay, were performed to characterize the response of the cells to the combination treatment. Results Combining 1,000 V/cm with 5 mmol/L Met yielded cell viability as low as 42.6% at 24 h. The glucose level was reduced by 5.60-fold and the ROS levels were increased by 9.56-fold compared to the control, leading to apoptotic cell death. Conclusions The results indicate the enhanced anticancer effect of Met in the presence of electric pulses. The cell growth is inhibited by suppressing glucose levels and elevated ROS. This shows a synergistic interplay between electroporation, Met, glucose, and ROS metabolic alterations. The results show promises for combinational therapy in TNBC patients.
Collapse
Affiliation(s)
- Praveen Sahu
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Ignacio G. Camarillo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Raji Sundararajan
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
275
|
Lu JX, Wang W, Zhang QW, Guo ZY, Jin Z, Tang YZ. Design, synthesis and evaluation of antioxidant and anti-inflammatory activities of novel resveratrol derivatives as potential multifunctional drugs. Eur J Med Chem 2024; 266:116148. [PMID: 38237344 DOI: 10.1016/j.ejmech.2024.116148] [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: 09/19/2023] [Revised: 12/25/2023] [Accepted: 01/10/2024] [Indexed: 02/05/2024]
Abstract
Oxidative stress and inflammation responses are closely related to the occurrence and development of many diseases. Therefore, anti-oxidation and anti-inflammation have become hot spots in the treatment of diseases. A series of novel resveratrol derivatives which hybrid with benzoylhydrazines were designed, synthesized and assessed for their in vitro antioxidant and anti-inflammatory activity. Initially, the antioxidant abilities of resveratrol derivatives were investigated by DPPH, ABTS radical scavenging and FRAP assays. RAW 264.7 macrophages are routinely used to evaluate the antioxidant and anti-inflammatory activities of drugs, so we used it to construct cell models of oxidative stress and inflammation. Among all the derivatives, compound 5 exhibited superior ROS- and NO-inhibitory activities. The molecular mechanism detected by Western blotting showed that compound 5 could significantly activate the Nrf2 signaling pathway and up-regulate the expression of HO-1 to resist oxidative stress stimulated by H2O2. At the same time, it could down-regulate the expression of apoptosis-related proteins Caspase3 and PARP, alleviating cells damage and apoptosis. In addition, compound 5 dose-dependently inhibited the activation of NF-κB p65/iNOS and MAPKs signaling pathway.
Collapse
Affiliation(s)
- Jia-Xun Lu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Wei Wang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Qi-Wen Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Zheng-Yan Guo
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Zhen Jin
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - You-Zhi Tang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
276
|
Li S, Fan R, Wang Y, He K, Xu J, Li H. Application of calcium overload-based ion interference therapy in tumor treatment: strategies, outcomes, and prospects. Front Pharmacol 2024; 15:1352377. [PMID: 38425645 PMCID: PMC10902152 DOI: 10.3389/fphar.2024.1352377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
Low selectivity and tumor drug resistance are the main hinderances to conventional radiotherapy and chemotherapy against tumor. Ion interference therapy is an innovative anti-tumor strategy that has been recently reported to induce metabolic disorders and inhibit proliferation of tumor cells by reordering bioactive ions within the tumor cells. Calcium cation (Ca2+) are indispensable for all physiological activities of cells. In particular, calcium overload, characterized by the abnormal intracellular Ca2+ accumulation, causes irreversible cell death. Consequently, calcium overload-based ion interference therapy has the potential to overcome resistance to traditional tumor treatment strategies and holds promise for clinical application. In this review, we 1) Summed up the current strategies employed in this therapy; 2) Described the outcome of tumor cell death resulting from this therapy; 3) Discussed its potential application in synergistic therapy with immunotherapy.
Collapse
Affiliation(s)
- Shuangjiang Li
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
- Battalion, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Ruicheng Fan
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Yuekai Wang
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
- Battalion, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Kunqian He
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
- Battalion, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Jinhe Xu
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Hongli Li
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
277
|
Lei Y, Chen Y, Wang S, Lin Z, Han P, Tian D, Wang H, Liu M. L-lysine supplementation attenuates experimental autoimmune hepatitis in a chronic murine model. Exp Anim 2024; 73:83-92. [PMID: 37648521 PMCID: PMC10877156 DOI: 10.1538/expanim.23-0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/22/2023] [Indexed: 09/01/2023] Open
Abstract
The incidence of autoimmune hepatitis (AIH) has increased significantly worldwide. The present study aims to explore the protective effect of L-lysine supplementation against AIH and to investigate its potential underlying mechanisms. A chronic experimental AIH mouse model was established by repeated tail vein injection of human cytochrome P450 2D6 (CYP2D6) plasmid. Starting from day 14 of the modeling, mice in the CYP2D6-AIH +L-lysine group were given 200 µl of purified water containing 10 mg/kg L-lysine by gavage until day27, once a day, and mice in the healthy control group and model group were given an equal volume of purified water by gavage. Our results showed that L-lysine supplementation partially reversed the liver injury mediated by CYP2D6 overexpression. These effects were consistent with the restraining impacts of L-lysine supplementation on decreasing pro-inflammatory cytokines expression level and CD4+ and CD8+ T lymphocytes infiltration, as well as curbing hepatic oxidative stress. Furthermore, L-lysine supplement relieved liver fibrosis in the context of AIH. In conclusion, L-lysine supplementation attenuates CYP2D6-induced immune liver injury in mice, which may serve as a novel nutrition support approach for AIH.
Collapse
Affiliation(s)
- Yu Lei
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, P.R. China
| | - Yu Chen
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, P.R. China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, P.R. China
| | - Zhuoying Lin
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, P.R. China
- Department of Gastroenterology, Shangrao People's Hospital, Shangrao 334000, Jiangxi Province, P.R. China
| | - Ping Han
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, P.R. China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, P.R. China
| | - Han Wang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, P.R. China
| | - Mei Liu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, P.R. China
| |
Collapse
|
278
|
Chang R, Han B, Ben Mabrouk A, Hasegawa U. Controlled Dissociation of Polymeric Micelles in Response to Oxidative Stress. Biomacromolecules 2024; 25:1162-1170. [PMID: 38227946 DOI: 10.1021/acs.biomac.3c01156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Nanoparticle-based drug carriers that can respond to oxidative stress in tumor tissue have attracted attention for site-specific drug release. Taking advantage of the characteristic microenvironment in tumors, one of the attractive directions in drug delivery research is to design drug carriers that release drugs upon oxidation. A strategy to incorporate oxidation-sensitive thioether motifs such as thiomorpholine acrylamide (TMAM) to drug carriers has been often used to achieve oxidation-induced dissociation, thereby targeted drug release. However, those delivery systems often suffer from a slow dissociation rate due to the intrinsic hydrophobicity of the thioether structures. In this study, we aimed to enhance the dissociation rate of TMAM-based micelles upon oxidation. The random copolymers of N-isopropylacrylamide and TMAM (P(NIPAM/TMAM)) were designed as an oxidation-sensitive segment that showed a fast response to oxidative stress. We first synthesized P(NIPAM/TMAM) copolymers with different NIPAM:TMAM molar ratios. Those copolymers exhibited low critical solution temperatures (LCSTs) below 32 °C, which shifted to higher temperatures after oxidation. The changes in LCSTs depend on the NIPAM:TMAM molar ratios. At the NIPAM:TMAM molar ratio of 82:18, the LCSTs before and after oxidation were 17 and 54 °C, respectively. We then prepared micelles from the diblock copolymers of poly(N-acryloyl morpholine) (PAM) and P(NIPAM/TMAM). The micelles showed an accelerated dissociation rate upon oxidation compared to the micelles without NIPAM units. Furthermore, the doxorubicin (Dox)-loaded micelles showed enhanced relative toxicity in human colorectal cancer (HT29) cells over human umbilical vein endothelial cells (HUVECs). Our novel strategy to design an oxidation-sensitive micellar core comprising a P(NIPAM/TMAM) segment can be used as a chemotherapeutic delivery system that responds to an oxidative tumor microenvironment in an appropriate time scale.
Collapse
Affiliation(s)
- Roujia Chang
- Department of Materials Science and Engineering, The Pennsylvania State University, Steidle Building, University Park, Pennsylvania 16802, United States
| | - Binru Han
- Department of Materials Science and Engineering, The Pennsylvania State University, Steidle Building, University Park, Pennsylvania 16802, United States
| | - Amira Ben Mabrouk
- Department of Materials Science and Engineering, The Pennsylvania State University, Steidle Building, University Park, Pennsylvania 16802, United States
| | - Urara Hasegawa
- Department of Materials Science and Engineering, The Pennsylvania State University, Steidle Building, University Park, Pennsylvania 16802, United States
| |
Collapse
|
279
|
Deng Y, Hou Z, Li Y, Yi M, Wu Y, Zheng Y, Yang F, Zhong G, Hao Q, Zhai Z, Wang M, Ma X, Kang H, Ji F, Dong C, Liu H, Dai Z. Superbinder based phosphoproteomic landscape revealed PRKCD_pY313 mediates the activation of Src and p38 MAPK to promote TNBC progression. Cell Commun Signal 2024; 22:115. [PMID: 38347536 PMCID: PMC10860301 DOI: 10.1186/s12964-024-01487-z] [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: 11/26/2023] [Accepted: 01/11/2024] [Indexed: 02/15/2024] Open
Abstract
Phosphorylation proteomics is the basis for the study of abnormally activated kinase signaling pathways in breast cancer, which facilitates the discovery of new oncogenic agents and drives the discovery of potential targets for early diagnosis and therapy of breast cancer. In this study, we have explored the aberrantly active kinases in breast cancer development and to elucidate the role of PRKCD_pY313 in triple negative breast cancer (TNBC) progression. We collected 47 pairs of breast cancer and paired far-cancer normal tissues and analyzed phosphorylated tyrosine (pY) peptides by Superbinder resin and further enriched the phosphorylated serine/threonine (pS/pT) peptides using TiO2 columns. We mapped the kinases activity of different subtypes of breast cancer and identified PRKCD_pY313 was upregulated in TNBC cell lines. Gain-of-function assay revealed that PRKCD_pY313 facilitated the proliferation, enhanced invasion, accelerated metastasis, increased the mitochondrial membrane potential and reduced ROS level of TNBC cell lines, while Y313F mutation and low PRKCD_pY313 reversed these effects. Furthermore, PRKCD_pY313 significantly upregulated Src_pY419 and p38_pT180/pY182, while low PRKCD_pY313 and PRKCD_Y313F had opposite effects. Dasatinib significantly inhibited the growth of PRKCD_pY313 overexpression cells, and this effect could be enhanced by Adezmapimod. In nude mice xenograft model, PRKCD_pY313 significantly promoted tumor progression, accompanied by increased levels of Ki-67, Bcl-xl and Vimentin, and decreased levels of Bad, cleaved caspase 3 and ZO1, which was opposite to the trend of Y313F group. Collectively, the heterogeneity of phosphorylation exists in different molecular subtypes of breast cancer. PRKCD_pY313 activates Src and accelerates TNBC progression, which could be inhibited by Dasatinib.
Collapse
Affiliation(s)
- Yujiao Deng
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhanwu Hou
- Center for Mitochondrial Biology and Medicine & Douglas C. Wallace Institute for Mitochondrial and Epigenetic Information Sciences, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yizhen Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Wu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Zheng
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fei Yang
- Center for Mitochondrial Biology and Medicine & Douglas C. Wallace Institute for Mitochondrial and Epigenetic Information Sciences, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guansheng Zhong
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qian Hao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhen Zhai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Wang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fanpu Ji
- Department of Infectious Diseases, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an, China
| | - Chenfang Dong
- Department of Pathology and Pathophysiology, Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Huadong Liu
- Center for Mitochondrial Biology and Medicine & Douglas C. Wallace Institute for Mitochondrial and Epigenetic Information Sciences, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
280
|
Tonon G. Myeloma and DNA damage. Blood 2024; 143:488-495. [PMID: 37992215 DOI: 10.1182/blood.2023021384] [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: 09/07/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/24/2023] Open
Abstract
ABSTRACT DNA-damaging agents have represented the first effective treatment for the blood cancer multiple myeloma, and after 65 years since their introduction to the clinic, they remain one of the mainstay therapies for this disease. Myeloma is a cancer of plasma cells. Despite exceedingly slow proliferation, myeloma cells present extended genomic rearrangements and intense genomic instability, starting at the premalignant stage of the disease. Where does such DNA damage stem from? A reliable model argues that the powerful oncogenes activated in myeloma as well the phenotypic peculiarities of cancer plasma cells, including the dependency on the proteasome for survival and the constant presence of oxidative stress, all converge on modulating DNA damage and repair. Beleaguered by these contraposing forces, myeloma cells survive in a precarious balance, in which the robust engagement of DNA repair mechanisms to guarantee cell survival is continuously challenged by rampant genomic instability, essential for cancer cells to withstand hostile selective pressures. Shattering this delicate equilibrium has been the goal of the extensive use of DNA-damaging agents since their introduction in the clinic, now enriched by novel approaches that leverage upon synthetic lethality paradigms. Exploiting the impairment of homologous recombination caused by myeloma genetic lesions or treatments, it is now possible to design therapeutic combinations that could target myeloma cells more effectively. Furthermore, DNA-damaging agents, as demonstrated in solid tumors, may sensitize cells to immune therapies. In all, targeting DNA damage and repair remains as central as ever in myeloma, even for the foreseeable future.
Collapse
Affiliation(s)
- Giovanni Tonon
- Università Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology and Center for Omics Sciences, Functional Genomics of Cancer Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
281
|
Fu Q, Yang X, Wang M, Zhu K, Wang Y, Song J. Activatable Probes for Ratiometric Imaging of Endogenous Biomarkers In Vivo. ACS NANO 2024; 18:3916-3968. [PMID: 38258800 DOI: 10.1021/acsnano.3c10659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Dynamic variations in the concentration and abnormal distribution of endogenous biomarkers are strongly associated with multiple physiological and pathological states. Therefore, it is crucial to design imaging systems capable of real-time detection of dynamic changes in biomarkers for the accurate diagnosis and effective treatment of diseases. Recently, ratiometric imaging has emerged as a widely used technique for sensing and imaging of biomarkers due to its advantage of circumventing the limitations inherent to conventional intensity-dependent signal readout methods while also providing built-in self-calibration for signal correction. Here, the recent progress of ratiometric probes and their applications in sensing and imaging of biomarkers are outlined. Ratiometric probes are classified according to their imaging mechanisms, and ratiometric photoacoustic imaging, ratiometric optical imaging including photoluminescence imaging and self-luminescence imaging, ratiometric magnetic resonance imaging, and dual-modal ratiometric imaging are discussed. The applications of ratiometric probes in the sensing and imaging of biomarkers such as pH, reactive oxygen species (ROS), reactive nitrogen species (RNS), glutathione (GSH), gas molecules, enzymes, metal ions, and hypoxia are discussed in detail. Additionally, this Review presents an overview of challenges faced in this field along with future research directions.
Collapse
Affiliation(s)
- Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Mengzhen Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Kang Zhu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
282
|
Li H, Wang J, Kim H, Peng X, Yoon J. Activatable Near-Infrared Versatile Fluorescent and Chemiluminescent Dyes Based on the Dicyanomethylene-4H-pyran Scaffold: From Design to Imaging and Theranostics. Angew Chem Int Ed Engl 2024; 63:e202311764. [PMID: 37855139 DOI: 10.1002/anie.202311764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 10/20/2023]
Abstract
Activatable fluorescent and chemiluminescent dyes with near-infrared emission have indispensable roles in the fields of bioimaging, molecular prodrugs, and phototheranostic agents. As one of the most popular fluorophore scaffolds, the dicyanomethylene-4H-pyran scaffold has been applied to fabricate a large number of versatile activatable optical dyes for analytes detection and diseases diagnosis and treatment by virtue of its high photostability, large Stokes shift, considerable two-photon absorption cross-section, and structural modifiability. This review discusses the molecular design strategies, recognition mechanisms, and both in vitro and in vivo bio-applications (especially for diagnosis and therapy of tumors) of activatable dicyanomethylene-4H-pyran dyes. The final section describes the current shortcomings and future development prospects of this topic.
Collapse
Affiliation(s)
- Haidong Li
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Jingyun Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Heejeong Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Korea
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Korea
| |
Collapse
|
283
|
Ge M, Papagiannakopoulos T, Bar-Peled L. Reductive stress in cancer: coming out of the shadows. Trends Cancer 2024; 10:103-112. [PMID: 37925319 DOI: 10.1016/j.trecan.2023.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 11/06/2023]
Abstract
Redox imbalance is defined by disruption in oxidative and reductive pathways and has a central role in cancer initiation, development, and treatment. Although redox imbalance has traditionally been characterized by high levels of oxidative stress, emerging evidence suggests that an overly reductive environment is just as detrimental to cancer proliferation. Reductive stress is defined by heightened levels of antioxidants, including glutathione and elevated NADH, compared with oxidized NAD, which disrupts central biochemical pathways required for proliferation. With the advent of new technologies that measure and manipulate reductive stress, the sensors and drivers of this overlooked metabolic stress are beginning to be revealed. In certain genetically defined cancers, targeting reductive stress pathways may be an effective strategy. Redox-based pathways are gaining recognition as essential 'regulatory hubs,' and a broader understanding of reductive stress signaling promises not only to reveal new insights into metabolic homeostasis but also potentially to transform therapeutic options in cancer.
Collapse
Affiliation(s)
- Maolin Ge
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA.
| | - Liron Bar-Peled
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
284
|
Liu X, Li P, Huang Y, Li H, Liu X, Du Y, Lin X, Chen D, Liu H, Zhou Y. M 6A demethylase ALKBH5 regulates FOXO1 mRNA stability and chemoresistance in triple-negative breast cancer. Redox Biol 2024; 69:102993. [PMID: 38104484 PMCID: PMC10770627 DOI: 10.1016/j.redox.2023.102993] [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: 11/03/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023] Open
Abstract
Resistance to chemotherapy is the main reason for treatment failure and poor prognosis in patients with triple-negative breast cancer (TNBC). Although the association of RNA N6-methyladenosine (m6A) modifications with therapy resistance is noticed, its role in the development of therapeutic resistance in TNBC is not well documented. This study aimed to investigate the potential mechanisms underlying reactive oxygen species (ROS) regulation in doxorubicin (DOX)-resistant TNBC. Here, we found that DOX-resistant TNBC cells displayed low ROS levels because of increased expression of superoxide dismutase (SOD2), thus maintaining cancer stem cells (CSCs) characteristics and DOX resistance. FOXO1 is a master regulator that reduces cellular ROS in DOX-resistant TNBC cells, and knockdown of FOXO1 significantly increased ROS levels by inhibiting SOD2 expression. Moreover, the m6A demethylase ALKBH5 promoted m6A demethylation of FOXO1 mRNA and increased FOXO1 mRNA stability in DOX-resistant TNBC cells. The analysis of clinical samples revealed that the increased expression levels of ALKBH5, FOXO1, and SOD2 were significantly positively correlated with chemoresistance and poor prognosis in patients with TNBC. To our knowledge, this is the first study to highlight that ALKBH5-mediated FOXO1 mRNA demethylation contributes to CSCs characteristics and DOX resistance in TNBC cells. Furthermore, pharmacological targeting of FOXO1 profoundly restored the response of DOX-resistant TNBC cells, both in vitro and in vivo. In conclusion, we demonstrated a critical function of ALKBH5-mediated m6A demethylation of FOXO1 mRNA in restoring redox balance, which in turn promoting CSCs characteristics and DOX resistance in TNBC, and suggested that targeting the ALKBH5/FOXO1 axis has therapeutic potential for patients with TNBC refractory to chemotherapy.
Collapse
Affiliation(s)
- Xi Liu
- Molecular Diagnosis Center, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China; Cancer Center Office, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Pan Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Yuanfeng Huang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Hongsheng Li
- Molecular Diagnosis Center, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Xin Liu
- Molecular Diagnosis Center, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Yaxi Du
- Molecular Diagnosis Center, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Xin Lin
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Danyang Chen
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Hao Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China.
| | - Yongchun Zhou
- Molecular Diagnosis Center, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China; Cancer Center Office, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China.
| |
Collapse
|
285
|
Pan L, Peng H, Lee B, Zhao J, Shen X, Yan X, Hua Y, Kim J, Kim D, Lin M, Zhang S, Li X, Yi X, Yao F, Qin Z, Du J, Chi Y, Nam JM, Hyeon T, Liu J. Cascade Catalytic Nanoparticles Selectively Alkalize Cancerous Lysosomes to Suppress Cancer Progression and Metastasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305394. [PMID: 37643367 DOI: 10.1002/adma.202305394] [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: 06/06/2023] [Revised: 08/16/2023] [Indexed: 08/31/2023]
Abstract
Lysosomes are critical in modulating the progression and metastasis for various cancers. There is currently an unmet need for lysosomal alkalizers that can selectively and safely alter the pH and inhibit the function of cancer lysosomes. Here an effective, selective, and safe lysosomal alkalizer is reported that can inhibit autophagy and suppress tumors in mice. The lysosomal alkalizer consists of an iron oxide core that generates hydroxyl radicals (•OH) in the presence of excessive H+ and hydrogen peroxide inside cancer lysosomes and cerium oxide satellites that capture and convert •OH into hydroxide ions. Alkalized lysosomes, which display impaired enzyme activity and autophagy, lead to cancer cell apoptosis. It is shown that the alkalizer effectively inhibits both local and systemic tumor growth and metastasis in mice. This work demonstrates that the intrinsic properties of nanoparticles can be harnessed to build effective lysosomal alkalizers that are both selective and safe.
Collapse
Affiliation(s)
- Limin Pan
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Haibao Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Bowon Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jiaxu Zhao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Xiulian Shen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ximei Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yipeng Hua
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jeonghyun Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dokyoon Kim
- Department of Bionano Engineering, Hanyang University, Ansan, 15588, Republic of Korea
| | - Mouhong Lin
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Shengjian Zhang
- Department of Radiology, Cancer Hospital/Institute and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaohui Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Xueying Yi
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Feibai Yao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yudan Chi
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Jwa-Min Nam
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jianan Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
286
|
Zhang J, Ye J, Zhu S, Han B, Liu B. Context-dependent role of SIRT3 in cancer. Trends Pharmacol Sci 2024; 45:173-190. [PMID: 38242748 DOI: 10.1016/j.tips.2023.12.005] [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: 08/28/2023] [Revised: 11/13/2023] [Accepted: 12/14/2023] [Indexed: 01/21/2024]
Abstract
Sirtuin 3 (SIRT3), an NAD+-dependent deacetylase, plays a key role in the modulation of metabolic reprogramming and regulation of cell death, as well as in shaping tumor phenotypes. Owing to its critical role in determining tumor-type specificity or the direction of tumor evolution, the development of small-molecule modulators of SIRT3, including inhibitors and activators, is of significant interest. In this review, we discuss recent studies on the oncogenic or tumor-suppressive functions of SIRT3, evaluate advances in SIRT3-targeted drug discovery, and present potential avenues for the design of small-molecule modulators of SIRT3 for cancer therapy.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shiou Zhu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
287
|
He Z, Feng D, Zhang C, Chen Z, Wang H, Hou J, Li S, Wei X. Recent strategies for evoking immunogenic Pyroptosis in antitumor immunotherapy. J Control Release 2024; 366:375-394. [PMID: 38142962 DOI: 10.1016/j.jconrel.2023.12.023] [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: 10/27/2023] [Revised: 11/24/2023] [Accepted: 12/16/2023] [Indexed: 12/26/2023]
Abstract
Pyroptosis is a specific type of programmed cell death (PCD) characterized by distinct morphological changes, including cell swelling, membrane blebbing, DNA fragmentation, and eventual cell lysis. Pyroptosis is closely associated with human-related diseases, such as inflammation and malignancies. Since the initial observation of pyroptosis in Shigella flexneri-infected macrophages more than 20 years ago, various pyroptosis-inducing agents, including ions, small molecules, and biological nanomaterials, have been developed for tumor treatment. Given that pyroptosis can activate the body's robust immune response against tumor and promote the formation of the body's long-term immune memory in tumor treatment, its status as a type of immunogenic cell death is self-evident. Therefore, pyroptosis should be used as a powerful anti-tumor strategy. However, there still is a lack of a comprehensive summary of the most recent advances in pyroptosis-based cancer therapy. Therefore, it is vital to fill this gap and inspire future drug design to better induce tumor cells to undergo pyroptosis to achieve advanced anti-tumor effects. In this review, we summarize in detail the most recent advances in triggering tumor cell immunogenic pyroptosis for adequate tumor clearance based on various treatment modalities, and highlight material design and therapeutic advantages. Besides, we also provide an outlook on the prospects of this emerging field in the next development.
Collapse
Affiliation(s)
- Zhangxin He
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215000, China
| | - Dexiang Feng
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Chaoji Zhang
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Zhiqian Chen
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - He Wang
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China.
| | - Jianquan Hou
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Shengliang Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215000, China.
| | - Xuedong Wei
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
288
|
Meng Y, Lin W, Wang N, Wei X, Mei P, Wang X, Zhang C, Huang Q, Liao Y. USP7-mediated ERβ stabilization mitigates ROS accumulation and promotes osimertinib resistance by suppressing PRDX3 SUMOylation in non-small cell lung carcinoma. Cancer Lett 2024; 582:216587. [PMID: 38097136 DOI: 10.1016/j.canlet.2023.216587] [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: 09/15/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2023]
Abstract
Osimertinib resistance is regarded as a major obstacle limiting survival benefits for patients undergoing treatment of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC). However, the underlying mechanisms of acquired resistance remain unclear. In this study, we report that estrogen receptor β (ERβ) is highly expressed in osimertinib-resistant NSCLC and plays a pivotal role in promoting osimertinib resistance. We further identified ubiquitin-specific protease 7 (USP7) as a critical binding partner that deubiquitinates and upregulates ERβ in NSCLC. ERβ promotes osimertinib resistance by mitigating reactive oxygen species (ROS) accumulation. We found that ERβ mechanistically suppresses peroxiredoxin 3 (PRDX3) SUMOylation and thus confers osimertinib resistance onto NSCLC. Furthermore, we provide evidence showing that depletion of ERβ induces ROS accumulation and reverses osimertinib resistance in NSCLC both in vitro and in vivo. Thus, our results demonstrate that USP7-mediated ERβ stabilization suppresses PRDX3 SUMOylation to mitigate ROS accumulation and promote osimertinib resistance, suggesting that targeting ERβ may be an effective therapeutic strategy to overcome osimertinib resistance in NSCLC.
Collapse
Affiliation(s)
- Yunchong Meng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Wei Lin
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Na Wang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Xiao Wei
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Peiyuan Mei
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Xiaojun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Chi Zhang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Quanfu Huang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| |
Collapse
|
289
|
Shi C, Zhang Y, Wu G, Zhu Z, Zheng H, Sun X, Heng Y, Pan S, Xiu H, Zhang J, Yin Z, Yu Z, Liang B. Hyaluronic Acid-Based Reactive Oxygen Species-Responsive Multifunctional Injectable Hydrogel Platform Accelerating Diabetic Wound Healing. Adv Healthc Mater 2024; 13:e2302626. [PMID: 37943252 DOI: 10.1002/adhm.202302626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/02/2023] [Indexed: 11/10/2023]
Abstract
Diabetic wounds are more likely to develop into complex and severe chronic wounds. The objective of this study is to develop and assess a reactive oxygen species (ROS)-responsive multifunctional injectable hydrogel for the purpose of diabetic wound healing. A multifunctional hydrogel (HA@Cur@Ag) is successfully synthesized with dual antioxidant, antibacterial, and anti-inflammatory properties by crosslinking thiol hyaluronic acid (SH-HA) and disulfide-bonded hyperbranched polyethylene glycol (HB-PBHE) through Michael addition; while, incorporating curcumin liposomes and silver nanoparticles (AgNPs). The HA@Cur@Ag hydrogel exhibits favorable biocompatibility, degradability, and injectivity. The outcomes of in vitro and in vivo experiments demonstrate that the hydrogel can effectively be loaded with and release curcumin liposomes, as well as silver ions, thereby facilitating diabetic wound healing through multiple mechanisms, including ROS scavenging, bactericidal activity, anti-inflammatory effects, and the promotion of angiogenesis. Transcriptome sequencing reveals that the HA@Cur@Ag hydrogel effectively suppresses the activation of the tumour necrosis factor (TNF)/nuclear factor κB (NF-κB) pathway to ameliorate oxidative stress and inflammation in diabetic wounds. These findings suggest that this ROS-responsive multifunctional injectable hydrogel, which possesses the ability to precisely coordinate and integrate intricate biological and molecular processes involved in wound healing, exhibits notable potential for expediting diabetic wound healing.
Collapse
Affiliation(s)
- Chen Shi
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, P. R. China
| | - Ying Zhang
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, P. R. China
| | - Guanfu Wu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, P. R. China
| | - Zhangyu Zhu
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, P. R. China
| | - Haiping Zheng
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, P. R. China
| | - Ximeng Sun
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, P. R. China
| | - Yongyuan Heng
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, P. R. China
| | - Shaowei Pan
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, P. R. China
| | - Haonan Xiu
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, P. R. China
| | - Jing Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, P. R. China
| | - Zhaowei Yin
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, P. R. China
| | - Ziyi Yu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing, 211816, P. R. China
| | - Bin Liang
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, P. R. China
| |
Collapse
|
290
|
Kim LC, Lesner NP, Simon MC. Cancer Metabolism under Limiting Oxygen Conditions. Cold Spring Harb Perspect Med 2024; 14:a041542. [PMID: 37848248 PMCID: PMC10835619 DOI: 10.1101/cshperspect.a041542] [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] [Indexed: 10/19/2023]
Abstract
Molecular oxygen (O2) is essential for cellular bioenergetics and numerous biochemical reactions necessary for life. Solid tumors outgrow the native blood supply and diffusion limits of O2, and therefore must engage hypoxia response pathways that evolved to withstand acute periods of low O2 Hypoxia activates coordinated gene expression programs, primarily through hypoxia inducible factors (HIFs), to support survival. Many of these changes involve metabolic rewiring such as increasing glycolysis to support ATP generation while suppressing mitochondrial metabolism. Since low O2 is often coupled with nutrient stress in the tumor microenvironment, other responses to hypoxia include activation of nutrient uptake pathways, metabolite scavenging, and regulation of stress and growth signaling cascades. Continued development of models that better recapitulate tumors and their microenvironments will lead to greater understanding of oxygen-dependent metabolic reprogramming and lead to more effective cancer therapies.
Collapse
Affiliation(s)
- Laura C Kim
- Abramson Family Cancer Research Institute, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Nicholas P Lesner
- Abramson Family Cancer Research Institute, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
291
|
Li Y, Yang C, Xie L, Shi F, Tang M, Luo X, Liu N, Hu X, Zhu Y, Bode AM, Gao Q, Zhou J, Fan J, Li X, Cao Y. CYLD induces high oxidative stress and DNA damage through class I HDACs to promote radiosensitivity in nasopharyngeal carcinoma. Cell Death Dis 2024; 15:95. [PMID: 38287022 PMCID: PMC10824711 DOI: 10.1038/s41419-024-06419-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024]
Abstract
Abnormal expression of Cylindromatosis (CYLD), a tumor suppressor molecule, plays an important role in tumor development and treatment. In this work, we found that CYLD binds to class I histone deacetylases (HDAC1 and HDAC2) through its N-terminal domain and inhibits HDAC1 activity. RNA sequencing showed that CYLD-HDAC axis regulates cellular antioxidant response via Nrf2 and its target genes. Then we revealed a mechanism that class I HDACs mediate redox abnormalities in CYLD low-expressing tumors. HDACs are central players in the DNA damage signaling. We further confirmed that CYLD regulates radiation-induced DNA damage and repair response through inhibiting class I HDACs. Furthermore, CYLD mediates nasopharyngeal carcinoma cell radiosensitivity through class I HDACs. Thus, we identified the function of the CYLD-HDAC axis in radiotherapy and blocking HDACs by Chidamide can increase the sensitivity of cancer cells and tumors to radiation therapy both in vitro and in vivo. In addition, ChIP and luciferase reporter assays revealed that CYLD could be transcriptionally regulated by zinc finger protein 202 (ZNF202). Our findings offer novel insight into the function of CYLD in tumor and uncover important roles for CYLD-HDAC axis in radiosensitivity, which provide new molecular target and therapeutic strategy for tumor radiotherapy.
Collapse
Affiliation(s)
- Yueshuo Li
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410078, China
- Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders/ Xiangya Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chenxing Yang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Longlong Xie
- Children's Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410008, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Min Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
- Molecular Imaging Research Center of Central South University, Changsha, 410008, Hunan, China
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
- Molecular Imaging Research Center of Central South University, Changsha, 410008, Hunan, China
| | - Na Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Xudong Hu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Yongwei Zhu
- Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders/ Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Qiang Gao
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Zhongshan Hospital, Shanghai Medical School, Fudan University, Shanghai, 200000, China
| | - Jian Zhou
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Zhongshan Hospital, Shanghai Medical School, Fudan University, Shanghai, 200000, China
| | - Jia Fan
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Zhongshan Hospital, Shanghai Medical School, Fudan University, Shanghai, 200000, China
| | - Xuejun Li
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders/ Xiangya Hospital, Central South University, Changsha, 410078, China.
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China.
- Molecular Imaging Research Center of Central South University, Changsha, 410008, Hunan, China.
- Department of Radiology, National Clinical Research Center for Geriatric Disorders/ Xiangya Hospital, Central South University, Changsha, 410078, China.
- Research Center for Technologies of Nucleic Acid-Based Diagnostics and Therapeutics Hunan Province, Changsha, 410078, China.
- National Joint Engineering Research Center for Genetic Diagnostics of Infectious Diseases and Cancer, Changsha, 410078, China.
| |
Collapse
|
292
|
Zhang J, Sun X, Chai X, Jiao Y, Sun J, Wang S, Yu H, Feng X. Curcumin Mitigates Oxidative Damage in Broiler Liver and Ileum Caused by Aflatoxin B1-Contaminated Feed through Nrf2 Signaling Pathway. Animals (Basel) 2024; 14:409. [PMID: 38338051 PMCID: PMC10854683 DOI: 10.3390/ani14030409] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
This experiment aimed to investigate the mitigating effect of CUR on the growth performance and liver and intestinal health of broilers fed AFB1-contaminated diets. In this study, 320 one-day-old healthy male Arbor Acres (AA) broilers were randomly divided into four groups, including the Control group (fed the basal diet), the AFB1 group (fed the AFB1-contaminated diet containing 1 mg/kg AFB1), the AFB1+CUR group (fed the AFB1-contaminated diet with 500 mg/kg CUR), and the CUR group (fed the basal diet containing 500 mg/kg CUR), with eight replicates of ten animals per group and a 28 d experimental period. In terms of the growth performance, the addition of 500 mg/kg CUR significantly improved AFB1-induced significant reductions in the final body weight on day 28 and mean daily gain (p < 0.05) and increased the ratio of the mean daily feed intake to mean daily weight gain in broilers (p < 0.05). In terms of liver health, significant improvements in liver histological lesions occurred in broilers in the AFB1+CUR group compared to the AFB1 group, with significantly higher glutathione peroxidase (GSH-Px), catalase (CAT), and total superoxide dismutase (T-SOD) activities (p < 0.05) and significantly higher levels of nuclear factor erythroid 2-related factor 2 (Nrf2), Kelch-like ECH-associated protein 1 (Keap-1), heme oxygenase 1 (HO-1), and NAD(P)H quinone oxidoreductase 1 (NQO-1) gene expression (p < 0.05). In terms of intestinal health, CUR addition significantly increased the relative length of ileum (p < 0.05), significantly elevated the height of ileal villi (p < 0.05), significantly reduced D-Lactate (D-LA) and diamine oxidase (DAO) activities in broiler serum (p < 0.05), significantly increased GSH, CAT, and T-SOD activities in ileal tissues (p < 0.05), and significantly elevated the expression of Nrf2, HO-1, and NQO-1 genes (p < 0.05) compared to the AFB1 group. In conclusion, CUR showed a protective effect against damage to the liver and intestine caused by AFB1 in broilers through the Nrf2 signaling pathway, thereby improving the growth performance of broilers exposed to AFB1.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xingjun Feng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (J.Z.); (X.S.); (X.C.); (Y.J.); (J.S.); (S.W.); (H.Y.)
| |
Collapse
|
293
|
Zhu W, Liu L, Wu J, Gao R, Fu L, Yang X, Zou Y, Zhang S, Luo D. SMYD3 activates the TCA cycle to promote M1-M2 conversion in macrophages. Int Immunopharmacol 2024; 127:111329. [PMID: 38091832 DOI: 10.1016/j.intimp.2023.111329] [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: 08/05/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND SMYD3 refers to a histone lysine methyltransferase from the SMYD family, which acts as a gene transcriptional regulator chiefly through catalysis of the histone subunit 3 at lysine 4 trimethylation (H3K4me3). Great progress has been made that epigenetic modification plays a pivotal role in regulating macrophage polarization. However, the effects of the histone lysine methyltransferase SMYD3 on macrophage polarization and phenotypic switching are unclear. RESULTS We found that LPS/IFN-γ-stimulated macrophages gradually transformed from M1 to M2 in the late stage, and SMYD3 played a key role in this process. As demonstrated by RNA-seq assessment, SMYD3 prominently activated a metabolic pathway known as TCA cycle inside macrophages during M1-M2 conversion. Besides, by modifying H3K4me3 histone, the target genes regulated by SMYD3 were identified via the ChIP-seq assessment, including citrate synthase (CS), succinate dehydrogenase complex subunit C (SDHC) and pyruvate carboxylase (PC). SMYD3 activated the transcriptional activities of the metabolic enzymes CS, SDHC and PC through H3K4me3 by causing the aggregation of citrate, an intramacrophage metabolite, and the depletion of succinate. And additionally, it facilitated the generation of ROS, as well as the expressions of genes associated with mitochondrial respiratory chain complexes. This increased ROS production ultimately induced mitophagy, triggering the M1 to M2 phenotype switch in the macrophages. CONCLUSIONS Our study provides a detailed intrinsic mechanism in the macrophage phenotypic transition process, in short, SMYD3 promotes the M1-M2 conversion of macrophages by activating the TCA cycle through the simultaneous regulation of the transcriptional activities of the metabolic enzymes CS, SDHC and PC.
Collapse
Affiliation(s)
- Wenqiang Zhu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Lina Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Jinjing Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Renzhuo Gao
- Queen Marry College, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Liying Fu
- Queen Marry College, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Xiaohong Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Yang Zou
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China
| | - Shuhua Zhang
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
294
|
Liu S, Shao F, Wang Y, Zhang Y, Yu H, Zhang N, He L, Kong Q, Jiang H, Dong Z. COX6C expression driven by copy amplification of 8q22.2 regulates cell proliferation via mediation of mitosis by ROS-AMPK signaling in lung adenocarcinoma. Cell Death Dis 2024; 15:74. [PMID: 38242874 PMCID: PMC10799076 DOI: 10.1038/s41419-024-06443-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/21/2024]
Abstract
Copy number variations (CNVs) play a vital role in regulating genes expression and tumorigenesis. We explored the copy number alterations in early-stage lung adenocarcinoma using high-throughput sequencing and nucleic acid flight mass spectrometry technology, and found that 8q22.1-22.2 is frequently amplified in lung adenocarcinoma tissues. COX6C localizes on the region and its expression is notably enhanced that driven by amplification in lung adenocarcinoma. Knockdown of COX6C significantly inhibits the cell proliferation, and induces S-G2/M cell cycle arrest, mitosis deficiency and apoptosis. Moreover, COX6C depletion causes a deficiency in mitochondrial fusion, and impairment of oxidative phosphorylation. Mechanistically, COX6C-induced mitochondrial deficiency stimulates ROS accumulation and activates AMPK pathway, then leading to abnormality in spindle formation and chromosome segregation, activating spindle assemble checkpoint, causing mitotic arrest, and ultimately inducing cell apoptosis. Collectively, we suggested that copy amplification-mediated COX6C upregulation might serves as a prospective biomarker for prognosis and targeting therapy in patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Shuanghui Liu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, P. R. China
| | - Fanggui Shao
- Department of Laboratory Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, P. R. China
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, P. R. China
| | - Yourong Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
- Department of clinical laboratory, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, P. R. China
| | - Yurui Zhang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Hongjia Yu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Ningxin Zhang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Lan He
- School of Biomedical Science, Hunan University, Changsha, Hunan, 410013, P. R. China
| | - Qingran Kong
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China.
| | - Hao Jiang
- Department of Biomedical Informatics, School of Life Sciences, Central South University, Changsha, Hunan, 410013, P. R. China.
| | - Zhixiong Dong
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China.
- Department of Laboratory Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, P. R. China.
| |
Collapse
|
295
|
Qian Z, Zhao N, Xu S, Yuan W. In situ injectable thermoresponsive nanocomposite hydrogel based on hydroxypropyl chitosan for precise synergistic calcium-overload, photodynamic and photothermal tumor therapy. Carbohydr Polym 2024; 324:121487. [PMID: 37985082 DOI: 10.1016/j.carbpol.2023.121487] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/21/2023] [Accepted: 10/10/2023] [Indexed: 11/22/2023]
Abstract
Traditional therapies have poor accuracy and significant toxic side effects in the process of tumor treatment. The non-traditional treatment methods with high accuracy and efficacy are worth exploring and investigating. Herein, a strategy that enables precise and synergistic therapies of calcium-overload, photodynamic, and photothermal through facile near infrared (NIR) irradiation was carried out base on the injectable and self-healable hydrogel encapsulating indocyanine green (ICG)-loaded and bovine serum albumin (BSA)-modified calcium peroxide (CaO2) nanoparticles (ICG@CaO2-BSA NPs) and bismuth sulfide (Bi2S3) nanorods. The hydrogel fabricated through the dynamic Schiff-base bonds between hydroxypropyl chitosan (HPCS) and aldehyde-modified Pluronic F127 (F127-CHO) as the delivery substrate for functional substances could adhere and grip tumor tissues due to the adhesion of hydroxyl groups in HPCS and the hydrophobic aggregation caused by thermoresponsiveness of F127-CHO. CaO2 in ICG@CaO2-BSA NPs decomposed in the tumor micro-acidic environment to produce calcium ions (Ca2+) and hydrogen peroxide (H2O2), while ICG generated reactive oxygen species (ROS) under NIR irradiation, the photothermal effect of Bi2S3 nanorods and ICG under NIR irradiation could increase the temperature of tumor tissues and ultimately achieve precise tumor cell destruction. Therefore, this strategy will provide promising prospects for precise and efficient treatment of tumors.
Collapse
Affiliation(s)
- Zhiyi Qian
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Nuoya Zhao
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Sicheng Xu
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Weizhong Yuan
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China.
| |
Collapse
|
296
|
Jiang Z, He J, Zhang B, Wang L, Long C, Zhao B, Yang Y, Du L, Luo W, Hu J, Hong X. A Potential "Anti-Warburg Effect" in Circulating Tumor Cell-mediated Metastatic Progression? Aging Dis 2024:AD.2023.1227. [PMID: 38300633 DOI: 10.14336/ad.2023.1227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
Metabolic reprogramming is a defining hallmark of cancer metastasis, warranting thorough exploration. The tumor-promoting function of the "Warburg Effect", marked by escalated glycolysis and restrained mitochondrial activity, is widely acknowledged. Yet, the functional significance of mitochondria-mediated oxidative phosphorylation (OXPHOS) during metastasis remains controversial. Circulating tumor cells (CTCs) are considered metastatic precursors that detach from primary or secondary sites and harbor the potential to seed distant metastases through hematogenous dissemination. A comprehensive metabolic characterization of CTCs faces formidable obstacles, including the isolation of these rare cells from billions of blood cells, coupled with the complexities of ex vivo-culturing of CTC lines or the establishment of CTC-derived xenograft models (CDX). This review summarized the role of the "Warburg Effect" in both tumorigenesis and CTC-mediated metastasis. Intriguingly, bioinformatic analysis of single-CTC transcriptomic studies unveils a potential OXPHOS dominance over Glycolysis signature genes across several important cancer types. From these observations, we postulate a potential "Anti-Warburg Effect" (AWE) in CTCs-a metabolic shift bridging primary tumors and metastases. The observed AWE could be clinically important as they are significantly correlated with therapeutic response in melanoma and prostate patients. Thus, unraveling dynamic metabolic regulations within CTC populations might reveal an additional layer of regulatory complexities of cancer metastasis, providing an avenue for innovative anti-metastasis therapies.
Collapse
Affiliation(s)
- Zhuofeng Jiang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Cancer Research Institute, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen Third People's Hospital, National Clinical Research Center for Infectious Diseases, Shenzhen, China
| | - Jiapeng He
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Cancer Research Institute, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen Third People's Hospital, National Clinical Research Center for Infectious Diseases, Shenzhen, China
| | - Binyu Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Cancer Research Institute, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen Third People's Hospital, National Clinical Research Center for Infectious Diseases, Shenzhen, China
| | - Liping Wang
- Department of Oncology, Southern University of Science and Technology Hospital, Shenzhen, Guangdong, China
| | - Chunhao Long
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Boxi Zhao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yufan Yang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Longxiang Du
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Weiren Luo
- Cancer Research Institute, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen Third People's Hospital, National Clinical Research Center for Infectious Diseases, Shenzhen, China
| | - Jianyang Hu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xin Hong
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
297
|
Wang J, Ma C, Tang Z, Sun Z, Qaed E, Chi X, Wang J, Jamalat Y, Geng Z, Tang Z, Yao Q. Mechanism study of oleanolic acid derivative, K73-03, inducing cell apoptosis in hepatocellular carcinoma. Cancer Cell Int 2024; 24:17. [PMID: 38185661 PMCID: PMC10771654 DOI: 10.1186/s12935-023-03119-x] [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: 08/09/2023] [Accepted: 10/30/2023] [Indexed: 01/09/2024] Open
Abstract
Oleanolic acid (3β-hydroxyolean-12-en-28-oic acid, OA) is a kind of pentacyclic triterpene, which widely distributes in nature. OA possesses a powerful anti-cancer effect; however, its low solubility limits its bioavailability and application. In this study, a new OA derivative, K73-03, was used to determine its effect on liver cancer cells and detailed molecular mechanisms. Here, we show that K73-03 may lead to the disorder of mitochondria in HepG2 cells, leading to excessive ROS production and apoptosis in cells. Meanwhile, K73-03 could induce cell apoptosis by inhibiting JAK2/STAT3 pathway and NF-κB/P65 pathway. Collectively, this study may provide a preliminary basis for further cancer treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of Physiology, Dalian Medical University, Dalian, China
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130000, China
| | - Chuchu Ma
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Zhongyuan Tang
- Department of Orthodontics, College of Stomatology, Jilin University, Changchun, Jilin, 130033, P.R. China
| | - Zhengwu Sun
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Eskandar Qaed
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Xinming Chi
- Histology and Embryology Department, Dalian Medical University, Dalian, China
| | - Jun Wang
- Pathophysiology Department, Dalian Medical University, Dalian, China
| | - Yazeed Jamalat
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Zhaohong Geng
- Department of Cardiology, 2th Affiliated Hospital of Dalian Medical University, Zhongshan Road No. 467, Dalian, 116000, China.
| | - Zeyao Tang
- Department of Pharmacology, Dalian Medical University, Dalian, China.
| | - Qiying Yao
- Department of Physiology, Dalian Medical University, Dalian, China.
| |
Collapse
|
298
|
Li J, Xu P, Chen S. Research progress on mitochondria regulating tumor immunity. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:1-14. [PMID: 38229501 PMCID: PMC10945498 DOI: 10.3724/zdxbyxb-2023-0484] [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: 10/08/2023] [Accepted: 12/01/2023] [Indexed: 01/18/2024]
Abstract
Tumor cells adapt their metabolism to meet the demands for energy and biosynthesis. Mitochondria, pivotal organelles in the metabolic reprogramming of tumor cells, contribute to tumorigenesis and cancer progression significantly through various dysfunctions in both tumor and immune cells. Alterations in mitochondrial dynamics and metabolic signaling pathways exert crucial regulatory influence on the activation, proliferation, and differentiation of immune cells. The tumor microenvironment orchestrates the activation and functionality of tumor-infiltrating immune cells by reprogramming mitochondrial metabolism and inducing shifts in mitochondrial dynamics, thereby facilitating the establishment of a tumor immunosuppressive microenvironment. Stress-induced leakage of mitochondrial DNA contributes multifaceted regulatory effects on anti-tumor immune responses and the immunosuppressive microenvironment by activating multiple natural immune signals, including cGAS-STING, TLR9, and NLRP3. Moreover, mitochondrial DNA-mediated immunogenic cell death emerges as a promising avenue for anti-tumor immunotherapy. Additionally, mitochondrial reactive oxygen species, a crucial factor in tumorigenesis, drives the formation of tumor immunosuppressive microenvironment by changing the composition of immune cells within the tumor microenvironment. This review focuses on the intrinsic relationship between mitochondrial biology and anti-tumor immune responses from multiple angles. We explore the core role of mitochondria in the dynamic interplay between the tumor and the host to facilitate the development of targeted mitochondrial strategies for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Jing Li
- College of Life and Environmental Science, Wenzhou University, Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, Wenzhou 325035, Zhejiang Province, China.
| | - Pinglong Xu
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China.
- Key Laboratory of Biosystem Homeostasis and Protection, Ministry of Education, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou 310058, China.
- Institute of Intelligent Medicine, Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311200, China.
- Cancer Center, Zhejiang University, Hangzhou 310058, China.
| | - Shasha Chen
- College of Life and Environmental Science, Wenzhou University, Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, Wenzhou 325035, Zhejiang Province, China.
| |
Collapse
|
299
|
Zhou Q, Zhang J, Zhang J, Liang S, Cai D, Xiao H, Zhu Y, Xiang W, Rodrigues-Lima F, Chi J, Guidez F, Wang L. Vemurafenib induces senescence in acute myeloid leukemia and myelodysplastic syndrome by activating the HIPPO signaling pathway: implications for potential targeted therapy. Biol Direct 2024; 19:6. [PMID: 38178263 PMCID: PMC10768477 DOI: 10.1186/s13062-023-00451-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND The outcome of Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) remain dismal despite the development of treatment. Targeted therapy is gaining more and more attention in improving prognosis. METHODS Expression of BRAF was analyzed by RT-qPCR in AML and MDS patients. Cells viability treated by drugs was measured by CCK-8 assay. Network pharmacology and RNA-sequence were used to analyze the mechanism of drugs and verified in vitro and xenograft tumor model. RESULTS Here we showed that BRAF was overexpressed in AML and MDS patients, and correlated with poor prognosis. The BRAF inhibitor-Vemurafenib (VEM) could significantly induce senescence, proliferation inhibition and apoptosis in AML cells, which can be enhanced by Bortezomib (BOR). This inhibitory effect was also verified in CD34 + cells derived from AML patients. Mechanistically, we showed that VEM combined with BOR could turn on HIPPO signaling pathway, thereby inducing cellular senescence in AML cells and xenograft mouse. CONCLUSIONS Taken together, our findings demonstrate a significant upregulation of BRAF expression in AML and MDS patients, which is associated with unfavorable clinical outcomes. We also discovered that the BRAF inhibitor Vemurafenib induces cellular senescence through activation of the HIPPO signaling pathway. Analysis of BRAF expression holds promise as a prognostic indicator and potential therapeutic target for individuals with AML and MDS.
Collapse
Affiliation(s)
- Qiao Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400000, People's Republic of China
| | - Jiamin Zhang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400000, People's Republic of China
| | - Jingsong Zhang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400000, People's Republic of China
| | - Simin Liang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400000, People's Republic of China
| | - Duo Cai
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400000, People's Republic of China
| | - Han Xiao
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400000, People's Republic of China
| | - Yu Zhu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400000, People's Republic of China
| | - Wenqiong Xiang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400000, People's Republic of China
| | - Fernando Rodrigues-Lima
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS UMR 8251, Paris, France
| | - Jianxiang Chi
- Center for the Study of Hematological Malignancies, Karaiskakio Foundation, Nicosia, Cyprus
| | - Fabien Guidez
- UMR1231 Inserm/uB/AgroSup, Université de Bourgogne, 7 boulevard Jeanne d'Arc 21079 DIJON Cedex, DIJON, France
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400000, People's Republic of China.
| |
Collapse
|
300
|
Todesca LM, Gerke M, Bulk EE, Bachmann M, Rudersdorf A, Antonuzzo L, Pillozzi S, Düfer M, Szabo I, Schwab A. Targeting K Ca3.1 channels to overcome erlotinib resistance in non-small cell lung cancer cells. Cell Death Discov 2024; 10:2. [PMID: 38177097 PMCID: PMC10767088 DOI: 10.1038/s41420-023-01776-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Almost all non-small cell lung cancer (NSCLC) patients initially responding to EGFR tyrosine kinase inhibitors (TKIs) develop acquired resistance. Since KCa3.1 channels, expressed in mitochondria and plasma membrane, regulate similar behavioral traits of NSCLC cells as EGFR, we hypothesized that their blockade contributes to overcoming EGFR-TKI resistance. Meta-analysis of microarray data revealed that KCa3.1 channel expression in erlotinib-resistant NSCLC cells correlates with that of genes of integrin and apoptosis pathways. Using erlotinib-sensitive and -resistant NSCLC cells we monitored the role of mitochondrial KCa3.1 channels in integrin signaling by studying cell-matrix adhesion with single-cell force spectroscopy. Apoptosis was quantified with fluorescence-based assays. The function of mitochondrial KCa3.1 channels in these processes was assessed by measuring the mitochondrial membrane potential and by quantifying ROS production. Functional assays were supplemented by biochemical analyses. We show that KCa3.1 channel inhibition with senicapoc in erlotinib-resistant NSCLC cells increases cell adhesion by increasing β1-integrin expression, that in turn depends on mitochondrial ROS release. Increased adhesion impairs migration of NSCLC cells in a 3D matrix. At the same time, the senicapoc-dependent ROS production induces cytochrome C release and triggers apoptosis of erlotinib-resistant NSCLC cells. Thus, KCa3.1 channel blockade overcomes EGFR-TKI resistance by inhibiting NSCLC motility and inducing apoptosis.
Collapse
Affiliation(s)
| | - Matthias Gerke
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Emma Etmar Bulk
- Institute of Physiology II, University of Münster, Münster, Germany
| | | | - Alisa Rudersdorf
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Lorenzo Antonuzzo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Martina Düfer
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Ildiko Szabo
- Department of Biology, University of Padova, Padua, Italy
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Münster, Germany
| |
Collapse
|