1
|
Wu Y, Yang L, You J, Tian C, Yang S, Li L. Discovery of phenazine derivatives as a new class of non-classical ferroptosis inhibitors and efficacy evaluation on a mouse model of liver injury. Eur J Med Chem 2025; 282:117042. [PMID: 39556893 DOI: 10.1016/j.ejmech.2024.117042] [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: 08/31/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024]
Abstract
Ferroptosis is an iron-dependent regulated cell death, which has been implicated in the onset and progression of numerous diseases. Ferroptosis inhibitors are thought as potential agents for treating these related diseases. However, the majority of currently available ferroptosis inhibitors are antioxidants or iron chelators (called classical ferroptosis inhibitors), which might have potential risks of side effects during clinical use. Herein, we report the discovery of phenazine derivatives as a new class of non-classical ferroptosis inhibitors. Structure-activity relationship of these series compounds led to the discovery of the most active compound 13l with an EC50 value of 0.0007 μM. Mechanistically, 13l could inhibit NCOA4-mediated ferritinophagy, hence protecting cells from ferroptosis. Notably, in the acetaminophen-induced acute liver injury model, 13l showed an excellent therapeutic effect. Overall, this compound reported here could be a promising lead compound for drug discovery targeting ferroptosis.
Collapse
Affiliation(s)
- Yunjie Wu
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Sichuan, 610041, China
| | - Lu Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Sichuan, 610041, China
| | - Jing You
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chenyu Tian
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shengyong Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Linli Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Sichuan, 610041, China.
| |
Collapse
|
2
|
Jalali-Zefrei F, Mousavi SM, Delpasand K, Shourmij M, Farzipour S. Role of Non-coding RNAs on the Radiotherapy Sensitivity and Resistance in Cancer Cells. Curr Gene Ther 2025; 25:113-135. [PMID: 38676526 DOI: 10.2174/0115665232301727240422092311] [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: 12/31/2023] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 04/29/2024]
Abstract
Radiotherapy (RT) is an integral part of treatment management in cancer patients. However, one of the limitations of this treatment method is the resistance of cancer cells to radiotherapy. These restrictions necessitate the introduction of modalities for the radiosensitization of cancer cells. It has been shown that Noncoding RNAs (ncRNAs), along with modifiers, can act as radiosensitivity and radioresistant regulators in a variety of cancers by affecting double strand break (DSB), wnt signaling, glycolysis, irradiation induced apoptosis, ferroptosis and cell autophagy. This review will provide an overview of the latest research on the roles and regulatory mechanisms of ncRNA after RT in in vitro and preclinical researches.
Collapse
Affiliation(s)
- Fatemeh Jalali-Zefrei
- Department of Cardiology, Cardiovascular Diseases Research Center, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyed Mehdi Mousavi
- Department of Cardiology, Cardiovascular Diseases Research Center, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Kourosh Delpasand
- Razi Clinical Research Development Unit, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Shourmij
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Soghra Farzipour
- Department of Cardiology, Cardiovascular Diseases Research Center, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
3
|
Fan L, Guo D, Zhu C, Gao C, Wang Y, Yin F, Liu M, Zhou Y, Wei T, Xiong X, Yu K, Le A. LRRC45 accelerates bladder cancer development and ferroptosis inhibition via stabilizing NRF2 by competitively KEAP1 interaction. Free Radic Biol Med 2025; 226:29-42. [PMID: 39522565 DOI: 10.1016/j.freeradbiomed.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Centrosomal dysregulation is closely linked to the genesis and progression of tumors. A comprehensive analysis of single-cell RNA sequencing (scRNA-seq) data has revealed that leucine-rich repeat-containing protein 45 (LRRC45), a centrosome linker protein crucial for maintaining centrosome cohesion and a member of the leucine-rich repeat-containing proteins (LRRCs) family, is significantly upregulated in bladder cancer. Notably, the elevated expression levels of LRRC45 were strongly correlated with a poor prognosis in patients. Furthermore, the depletion of LRRC45 in bladder cancer cells markedly inhibited tumorigenic proliferation and increased intracellular iron and reactive oxygen species (ROS) levels. It ultimately triggered ferroptosis, an iron-dependent form of programmed cell death characterized by lipid peroxidation. Mechanistic studies revealed that LRRC45 exerts its oncogenic effects through competitive interaction with Kelch-like ECH-associated protein 1 (KEAP1), which inhibits the ubiquitin-proteasome-mediated degradation of nuclear factor erythroid 2-related factor 2 (NRF2). This interaction enhances the nuclear translocation of NRF2 and its subsequent anti-ferroptotic activity. In conclusion, our studies highlight the critical role of LRRC45 in enhancing the stability of NRF2, thereby promoting the tumorigenic potential of bladder cancer. These insights suggest that targeting LRRC45 could serve as a promising molecular target for developing novel therapeutic interventions for bladder cancer.
Collapse
Affiliation(s)
- Linwei Fan
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Dingfan Guo
- The First Clinical Medical School of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chao Zhu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Chenqi Gao
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yu Wang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Fang Yin
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Mengwei Liu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yanyu Zhou
- The First Clinical Medical School of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Tiancheng Wei
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Xinxin Xiong
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Kuai Yu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Aiping Le
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
4
|
Ma S, Xu M, Zhang J, Li T, Zhou Q, Xi Z, Wang Z, Wang J, Ge Y. Analysis and functional validations of multiple cell death patterns for prognosis in prostate cancer. Int Immunopharmacol 2024; 143:113216. [PMID: 39353397 DOI: 10.1016/j.intimp.2024.113216] [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: 05/28/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
Prostate cancer (PCa) has garnered significant attention due to its rising incidence, variable therapeutic outcomes, and the absence of reliable prognostic markers. The significance of different cell death patterns in tumor development underscores their potential as predictors of PCa prognosis. This study utilized The Cancer Genome Atlas (TCGA) datasets to evaluate the prognostic capabilities of 15 cell death patterns and established a Cell Death Index (CDI) signature based on necrosis and cuproptosis-related genes. The predictive efficacy of the CDI signature was validated in our PCa cohort and in two public datasets: Deutsches Krebsforschungszentrum (DKFZ) and Memorial Sloan-Kettering Cancer Center (MSKCC) PCa cohorts. Our comprehensive analysis examined the relationship between CDI signature and clinical characteristics, published prognostic signatures, gene mutations, immune cell infiltration, enrichment pathways, and drug sensitivity in PCa. In vitro and in vivo studies assessed the impact of EDA2R and LOXL2 on PCa progression. The CDI signature exhibited robust predictive performance across three independent validation sets, with 1-, 2-, 3-, 4-, and 5-year area under the curve (AUC) values in the TCGA cohort of 0.866, 0.77, 0.836, 0.776, and 0.787, respectively. Higher CDI scores were correlated with advanced T and N stages, elevated Gleason scores, increased immune cell infiltration, gene mutations, and drug sensitivity. EDA2R inhibited PCa cell proliferation and migration, related to tumor necrosis, while LOXL2 promoted these processes and was associated with cuproptosis. In summary, our study identified a novel CDI signature as an effective indicator for diagnosis, personalized treatment, and prognostic assessment in PCa.
Collapse
Affiliation(s)
- Sheng Ma
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Mengyao Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Junbiao Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tengfei Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Qiang Zhou
- Department of Urology, Qinghai University Affiliated Hospital, Qinghai University Medical College, Xining, Qinghai 810001, China
| | - Zirui Xi
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Jing Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Yue Ge
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
5
|
Ma M, Hu R, Huang Q, Li J, Lv M, Sun J, Zhong X, Yi J, Peng L, Feng W, Ma W, Han Z, Zhang W, Sun X, Zhan B, Liu X, Zhou X. Digoxigenin activates autophagy in hepatocellular carcinoma cells by regulating the PI3K/AKT/mTOR pathway. Cancer Cell Int 2024; 24:405. [PMID: 39696358 DOI: 10.1186/s12935-024-03602-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is recognized as a highly malignant tumor. Targeted combination immunotherapy, the initially approved regimen, is compromised by adverse side effects and low response rates during clinical treatment. Traditional Chinese medicine and its derived natural compounds, known for their anticancer effects, offer advantages of low toxicity and cost. In this study, we performed high-throughput phenotypic screening in vitro to identify promising anti-HCC drugs. Among 1,444 bioactive compounds, digoxigenin (DIG) was found to significantly impede HCC cell progression. We validated DIG's therapeutic effects through assays such as cell counting by CCK8, lactate dehydrogenase, and colony formation. Analyses including transmission electron microscopy, western blotting, and immunofluorescence demonstrated that DIG inhibits HCC cell proliferation via autophagy. Network pharmacology and molecular docking studies suggest that DIG targets the PI3K/AKT/mTOR signaling pathway. Comparative treatments of Hep3B and Huh7 cells with DIG or mTOR inhibitors revealed similar inhibitory impacts, indicating that DIG induces autophagy by inhibiting the PI3K/AKT/mTOR pathway. In vivo studies confirmed that DIG halts the growth of subcutaneous xenograft tumors. In conclusion, DIG represents a potential HCC treatment by modulating the PI3K/AKT/mTOR pathway to induce autophagy. This research, via phenotypic screening, accelerates drug discovery and the development of novel therapies targeting the underlying mechanisms of liver cancer.
Collapse
Affiliation(s)
- Mengqing Ma
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Rui Hu
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Qi Huang
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Jing Li
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Minling Lv
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Jialing Sun
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Xin Zhong
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Jinyu Yi
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Lanfen Peng
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Wenxing Feng
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Wenfeng Ma
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Zhiyi Han
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Wei Zhang
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Xinfeng Sun
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Bolin Zhan
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Xingning Liu
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Xiaozhou Zhou
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China.
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China.
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China.
| |
Collapse
|
6
|
Xu C, Wen S, Du X, Zou X, Leung ELH, Zhou G, Wu Q, Shen B. Targeting regulated cell death (RCD) with naturally derived sesquiterpene lactones in cancer therapy. Pharmacol Res 2024:107553. [PMID: 39706282 DOI: 10.1016/j.phrs.2024.107553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Regulated cell death (RCD) is a type of cell death modulated by specific signal transduction pathways. Currently, known RCD types include apoptosis, autophagy, ferroptosis, necroptosis, cuproptosis, pyroptosis, and NETosis. Mutations in cancer cells may prevent the RCD pathway; therefore, targeting RCD in tumors has become a promising therapeutic approach. Sesquiterpene lactones represent a diverse and extensive class of plant-derived phytochemicals that serve as potential sources for developing various drugs. Recent studies have shown that sesquiterpene lactones have promising potential in cancer treatment. This review systematically summarizes recent progress in the study of sesquiterpene lactones as antitumor agents, highlighting their role in targeting various RCD pathways, including those involved in apoptosis, autophagy, ferroptosis, necroptosis, and cuproptosis. The primary purpose of the present review is to provide a clear picture of the regulation of RCD by sesquiterpene lactones against different targets in various cancers, which will facilitate the development of new strategies for cancer therapy.
Collapse
Affiliation(s)
- Cong Xu
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China; State Key Laboratory of Quality Research in Chinese Medicines and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Shaodi Wen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China
| | - Xiaoyue Du
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China
| | - Xinhua Zou
- Department of Vascular and Tumor Interventional Medicine, Affiliated Hospital of Jining Medical University, Jining 272000, China
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Science, University of Macau, 999078, Macao Special Administrative Region of China
| | - Guoren Zhou
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China.
| | - Bo Shen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China; DongTai People's Hospital, Dongtai, Jiangsu, China.
| |
Collapse
|
7
|
Uchimura R, Nishimura S, Ozaki M, Kurogi M, Kawahara K, Makise M, Kuniyasu A. Targeting Non-Apoptotic Pathways with the Cell Permeable TAT-Conjugated NOTCH1 RAM Fragment for Leukemia and Lymphoma Cells. ACS OMEGA 2024; 9:49925-49934. [PMID: 39713683 PMCID: PMC11656380 DOI: 10.1021/acsomega.4c08955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 12/24/2024]
Abstract
Targeting nonapoptotic cell death offers a promising strategy for overcoming apoptosis resistance in cancer. In this study, we developed Tat-Ram13, a 25-mer peptide that fuses the NOTCH1 intracellular domain fragment RAM13 with a cell-penetrating HIV-1 TAT, for the treatment of T-cell acute lymphoblastic leukemia with aberrant NOTCH1 mutation. Tat-Ram13 significantly downregulated NOTCH1-target genes in T-ALL cell lines. Furthermore, the peptide had potent cytotoxic effects on various human leukemia and lymphoma cell lines. However, it did not affect normal lymphocytes and monocytes, some subsets of leukemia cells, or adherent tumor cells. This cell-selective cytotoxic activity was closely correlated with the peptide uptake via macropinocytosis in leukemia cells. In leukemia cells, Tat-Ram13 triggered rapid cell death. This cell death involved mitochondrial membrane depolarization and extracellular release of lactate dehydrogenase and high-mobility group box-1 protein without activation of caspase-3 or cleavage of PARP-1. These results suggest that Tat-Ram13 cell death is nonapoptotic and mediated by rapid plasma membrane rupture. Moreover, alanine scanning analysis identified four critical hydrophobic amino acids in the RAM13 domain essential for its cytotoxicity. Consequently, these results suggest that Tat-Ram13 is a tumor-selective, nonapoptotic cell death-inducing agent for treating refractory leukemia and lymphomas with apoptosis resistance.
Collapse
Affiliation(s)
- Ryota Uchimura
- Faculty
of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Shinpei Nishimura
- Faculty
of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe, Chuo-ku, Kumamoto 862-0973, Japan
| | - Mikako Ozaki
- Faculty
of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Manami Kurogi
- Faculty
of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe, Chuo-ku, Kumamoto 862-0973, Japan
| | - Kohichi Kawahara
- Faculty
of Pharmacy, Niigata University of Pharmacy
and Applied Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata 956-0841, Japan
| | - Masaki Makise
- Faculty
of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Akihiko Kuniyasu
- Faculty
of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| |
Collapse
|
8
|
Tu S, Zou Y, Yang M, Zhou X, Zheng X, Jiang Y, Wang H, Chen B, Qian Q, Dou X, Bao J, Tian L. Ferroptosis in hepatocellular carcinoma: Mechanisms and therapeutic implications. Biomed Pharmacother 2024; 182:117769. [PMID: 39689515 DOI: 10.1016/j.biopha.2024.117769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024] Open
Abstract
Ferroptosis is a novel form of oxidative cell death, in which highly expressed unsaturated fatty acids on the cell membrane are catalyzed by divalent iron or ester oxygenase to promote liposome peroxidation. This process reduces cellular antioxidant capacity, increases lipid reactive oxygen species, and leads to the accumulation of intracellular ferrous ions, which disrupts intracellular redox homeostasis and ultimately causes oxidative cell death. Studies have shown that ferroptosis induces an immune response that has a dual role in liver disease, ferroptosis also offers a promising strategy for precise cancer therapy. Ferroptosis regulators are beneficial in maintaining cellular homeostasis and tissue health, have shown efficacy in treating diseases of the hepatic system. However, the mechanisms of action and molecular regulatory pathways of ferroptosis in hepatocellular carcinoma (HCC) have not been fully elucidated. Therefore, deciphering the role of ferroptosis and its mechanisms in HCC progression is crucial for treating the disease. In this review, we introduce the morphological features and biochemical functions of ferroptosis, outline the molecular regulatory pathways of ferroptosis, and highlights the therapeutic potential of ferroptosis inhibitors and modulators to target it in HCC.
Collapse
Affiliation(s)
- Shanjie Tu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yuchao Zou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Meiqi Yang
- Liaoning University of Traditional Chinese Medicine Xinglin College, Shenyang, Liaoning, PR China
| | - Xinlei Zhou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Xu Zheng
- The First Affiliated Hospital of Henan University of TCM, Zhengzhou, Henan, PR China
| | - Yuwei Jiang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Haoran Wang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Buyang Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Qianyu Qian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| | - Jianfeng Bao
- The Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| | - Lulu Tian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
9
|
Carlos A, Mendes M, Cruz MT, Pais A, Vitorino C. Ferroptosis driven by nanoparticles for tackling glioblastoma. Cancer Lett 2024:217392. [PMID: 39681210 DOI: 10.1016/j.canlet.2024.217392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Glioblastoma (GBM) is the most aggressive, malignant, and drug-resistant brain tumor. There are no effective treatment options for GBM, which usually leads to relapses that cause patients to die a few months later. Ferroptosis, a newly discovered mechanism of regulated cell death, has been identified as a tumor suppressor in solid tumors and represents an alternative to apoptosis resistance. This mechanism of cell death is characterized by iron overload, which is responsible for generating reactive oxygen species (ROS) in the cell. Understanding the ferroptosis pathway and its key regulators can be used to develop rational delivery systems that specifically target these regulators in GBM cells and promote cell death. This review conducted a systematic literature search to better understand the potential of ferroptosis as a target for developing nanoparticles to tackle GBM. The mechanisms of action, design parameters, efficacy, and safety concerns of 16 nanoparticles were evaluated, demonstrating the potential of combining ferroptosis inducers with nanocarriers to promote a selective delivery to the tumor microenvironment.
Collapse
Affiliation(s)
- Ana Carlos
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Faculty of Sciences and Technology, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Maria T Cruz
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal; Center for Neurosciences and Cell Biology (CNC) and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Alberto Pais
- Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Faculty of Sciences and Technology, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Faculty of Sciences and Technology, University of Coimbra, 3004-535, Coimbra, Portugal.
| |
Collapse
|
10
|
Wang X, Xie C, Lin L. Disulfidoptosis-related molecular subtypes and prognostic model for optimizing drug therapy in metastatic osteosarcoma patients. FASEB J 2024; 38:e70258. [PMID: 39673552 DOI: 10.1096/fj.202401510r] [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: 07/03/2024] [Revised: 09/24/2024] [Accepted: 11/04/2024] [Indexed: 12/16/2024]
Abstract
Disulfidotosis is a newly identified form of cell death associated with tumor response, patient outcomes, and cancer progression. This study aims to identify disulfidptosis-related genes (DiRGs) and their role in osteosarcoma (OS) to predict prognosis and optimize drug therapy for better patient survival. Gene expression matrices and clinical information on OS were obtained from the TARGET and GEO databases. Unsupervised clustering analysis identified two DiRG molecular subgroups with significantly different intratumoral heterogeneity and tumor microenvironment cell infiltrating characteristics in OS. A robust disulfidptosis-related prognostic model using five DiRGs was developed, demonstrating excellent predictive and prognostic power in OS with AUC values of 0.69, 0.78, and 0.85 for 1-, 3-, and 5-year periods, respectively. Investigations into the impact of disulfidoptosis on immune status in OS patients across different risk subgroups revealed that a low immune score and compromised immune status were associated with an unfavorable prognosis for OS patients. INF2 and MEGF10 genes are highly reliable predictors of metastasis among the hub DiRG genes. The validation results indicate a robust correlation between the expression of INF2 and MEGF10 and the severity of malignancy and metastasis in OS. Six drugs targeting osteosarcoma metastasis were identified, with INF2-BP-1-102 and MEGF10-AS703569 showing the best docking scores, indicating their potential to treat OS metastasis effectively. These findings provide valuable insight into improving treatment for OS patients.
Collapse
Affiliation(s)
- Xiaoping Wang
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
- Department of Orthopedics, Zhongshan Xiaolan People's Hospital (The Fifth People's Hospital of Zhongshan), Zhongshan, Guangdong, China
| | - Chao Xie
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Lijun Lin
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
11
|
Tao R, Li Y, Gong S, Zhang Q, Zhu Z. Unveiling intricating roles and mechanisms of ferroptosis in melanoma. Biochim Biophys Acta Rev Cancer 2024; 1880:189234. [PMID: 39644939 DOI: 10.1016/j.bbcan.2024.189234] [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: 06/06/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
Melanoma is a highly invasive malignant tumor originating from melanocytes, with increasing incidence in recent years. Ferroptosis is an iron-dependent and non-apoptotic form of programmed cell death characterized by the accumulation of lipid peroxides and reactive oxygen species. Morphologically, ferroptosis exhibits the alteration in cells, such as reduced mitochondrial volume, increased density of bilayer membrane, and a decrease or disappearance of mitochondrial cristae. Ferroptosis has shown tremendous potential and applicability in regulating the development of melanoma. As melanoma progresses, certain biomarkers associated with ferroptosis display characteristic patterns of expression. These changes not only reveal the sensitivity of tumor cells to ferroptosis but also provide potential targets for diagnosis and treatment. Besides, inducing ferroptosis has been well-documented to inhibit the growth of melanoma and enhance the efficacy of tumor immunotherapy. Hence, this review emphasizes the roles and regulatory mechanisms of ferroptosis in melanoma development, the involved immune regulation, as well as the potential for diagnosis and treatment of melanoma. The continuous explorations will endow novel strategies for developing ferroptosis-based therapies for melanoma.
Collapse
Affiliation(s)
- Rui Tao
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yichuan Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Song Gong
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China; Xianning Medical College, Hubei University of Science & Technology, Xianning 437000, Hubei Province, China.
| | - Zhanyong Zhu
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| |
Collapse
|
12
|
Hu J, Zhu J, Chen T, Zhao Y, Xu Q, Wang Y. Cuproptosis in cancer therapy: mechanisms, therapeutic application and future prospects. J Mater Chem B 2024; 12:12191-12206. [PMID: 39526989 DOI: 10.1039/d4tb01877j] [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: 11/16/2024]
Abstract
Cuproptosis is a regulated form of cell death induced by the accumulation of metal ions and is closely linked to aspects of cellular drug resistance, cellular metabolism, and signalling pathways. Due to its crucial role in regulating physiological and pathological processes, cuproptosis has gained increasing significance as a potential target for anticancer drug development. In this review, we introduce the definition of cuproptosis and provide a comprehensive discussion of the mechanisms of cuproptosis. In addition, the methods for the detection of cuproptosis are summarized, and recent advances in cuproptosis in cancer therapy are reviewed, mainly in terms of elesclomol (ES)-mediated cuproptosis and disulfiram (DSF)-mediated cuproptosis, which provided practical value for applications. Finally, the current challenges and future development of cuproptosis-mediated cancer therapy are discussed. In summary, this review highlights recent progress on cuproptosis in cancer therapy, offering novel ideas and strategies for future research and applications.
Collapse
Affiliation(s)
- Jiawei Hu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Junfei Zhu
- China-Japan Friendship Hospital, No. 2 Sakura East Street, Chaoyang District, Beijing, China
| | - Tao Chen
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yudie Zhao
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Qingwen Xu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| |
Collapse
|
13
|
Zhu H, Zhao C, Zhu H, Xu X, Hu C, Zhang Z. The characteristics and functional significance of disulfidptosis-related genes in head and neck squamous cell carcinoma. Discov Oncol 2024; 15:739. [PMID: 39625660 PMCID: PMC11615178 DOI: 10.1007/s12672-024-01629-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
Disulfidptosis is a newfound programmed cell death (PCD) mode characterized by disulfide stress. Nevertheless, the characteristics and functional significance of disulfidptosis-related genes in head and neck squamous cell carcinoma (HNSCC) are still largely unknown. In this study, several computer-aided bioinformatic analyses were performed. The Nonnegative Matrix Factorization (NMF) method classified The Cancer Genome Atlas (TCGA) patients into two clusters according to the expression of disulfidptosis-related genes. The relative compositions of cells in the tumor microenvironment (TME), mutant landscape, lasso regression analysis, and predicted clinical outcome were performed by analyzing bulk RNA-sequencing data. Besides, single-cell sequencing data (scRNA) was analyzed by Seurat, CopyKAT, and monocle2 to reveal the expression characteristics of disulfidptosis-related genes. Moreover, the spatial distribution characteristics of each cell subgroup in the section and the functional significance of cancer-associated fibroblasts (CAFs) were elucidated by STUtility, SpaCET, and SPATA2. Here, two clusters with different expression characteristics of disulfidptosis-related genes were identified. Cluster 1 (C1) patients had a worse prognosis and a higher proportion of stromal cells but lower effector T cell infiltration than cluster 2 (C2). A novel prognostic model was established and verified in our patient cohort. Additionally, diploid and inflammatory CAFs (iCAFs) showed higher disulfidptosis-related gene expression levels. Furthermore, the CCNC and CHMP1B expressions significantly changed following CAFs differentiation. Disulfidptosis-related genes exhibited extensive and differential spatial expression on tissue sections. Collectively, our study may contribute to revealing the function of disulfidptosis, and improve the expansion of knowledge of crosstalk between cancer cells and CAFs.
Collapse
Affiliation(s)
- Haiqian Zhu
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang Province, People's Republic of China
| | - Chifeng Zhao
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang Province, People's Republic of China
| | - Haoran Zhu
- Xi'an Jiaotong University Health Science Center, Xi'an, 710000, Shaanxi Province, China
| | - Xuhui Xu
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang Province, People's Republic of China
| | - Conglin Hu
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang Province, People's Republic of China
| | - Zhenxing Zhang
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), No.999, Donghai Avenue, Taizhou, 318000, Zhejiang Province, People's Republic of China.
| |
Collapse
|
14
|
Han L, Yang H, Jiang X, Zhou Z, Ge C, Yu K, Li G, Wang W, Liu Y. Prognostic model based on disulfidptosis-related lncRNAs for predicting survival and therapeutic response in bladder cancer. Front Immunol 2024; 15:1512203. [PMID: 39687628 PMCID: PMC11647029 DOI: 10.3389/fimmu.2024.1512203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Background With poor treatment outcomes and prognosis, bladder cancer remains a focus for clinical research in the precision oncology era. However, the potential of disulfidptosis, a novel cell death mechanism, and its related long non-coding RNAs to support selective cancer cell killing in this disease is still unclear. Methods We identified key disulfidptosis-related lncRNAs in bladder cancer, constructed a prognostic risk model with potential therapeutic targets, and confirmed the findings through quantitative PCR analysis. Results We identified five crucial lncRNAs (AC005840.4, AC010331.1, AL021707.6, MIR4435-2HG and ARHGAP5-AS1) and integrated them into a predictive model centered on disulfidptosis-associated lncRNAs. Reliability and validity tests demonstrated that the lncRNA prediction index associated with disulfidptosis effectively discerns patients' prognosis outcomes. Additionally, high-risk patients exhibited elevated expression levels of genes involved in the PI3K-Akt signaling pathway, extracellular matrix organization, and immune escape mechanisms, which are associated with poor prognosis. Notably, high-risk patients demonstrated higher sensitivity to Sorafenib, Oxaliplatin and MK-2206, underscoring the promise of these lncRNAs as precise therapeutic targets in bladder cancer. Conclusion By revealing the predictive importance of disulfidptosis-associated lncRNAs in bladder cancer, our research offers new perspectives and pinpoints potential therapeutic targets in clinical environments.
Collapse
Affiliation(s)
- Lirui Han
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Hankai Yang
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Xuan Jiang
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Ziyu Zhou
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Chang Ge
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Kairan Yu
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Guofang Li
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
| | - Wei Wang
- Ministry of Education (MOE) Key Laboratory of Bio-Intelligent Manufacturing, Dalian University of Technology, Dalian, China
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Liaoning, Shenyang, China
| | - Yubo Liu
- Department of Life and Pharmaceutical Sciences, School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, China
- Ministry of Education (MOE) Key Laboratory of Bio-Intelligent Manufacturing, Dalian University of Technology, Dalian, China
| |
Collapse
|
15
|
Chen J, Zhao L, Xu MF, Huang D, Sun XL, Zhang YX, Li HM, Wu CZ. Novel isobavachalcone derivatives induce apoptosis and necroptosis in human non-small cell lung cancer H1975 cells. J Enzyme Inhib Med Chem 2024; 39:2292006. [PMID: 38086769 DOI: 10.1080/14756366.2023.2292006] [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: 08/31/2023] [Accepted: 12/03/2023] [Indexed: 12/18/2023] Open
Abstract
In this study, seventeen isobavachalcone (IBC) derivatives (1-17) were synthesised, and evaluated for their cytotoxic activity against three human lung cancer cell lines. Among these derivatives, compound 16 displayed the most potent cytotoxic activity against H1975 and A549 cells, with IC50 values of 4.35 and 14.21 μM, respectively. Compared with IBC, compound 16 exhibited up to 4.11-fold enhancement of cytotoxic activity on human non-small cell lung cancer H1975 cells. In addition, we found that compound 16 suppressed H1975 cells via inducing apoptosis and necroptosis. The initial mechanism of compound 16 induced cell death in H1975 cells involves the increasing of Bax/Bcl-2 ratio and Cyt C protein level, down-regulating of Akt protein level, and cleaving caspase-9 and -3 induced apoptosis; the up-regulation of RIP3, p-RIP3, MLKL, and p-MLKL levels induced necroptosis. Moreover, compound 16 also caused mitochondrial dysfunction, thereby decreasing cellular ATP levels, and resulting in excessive reactive oxygen species (ROS) accumulation.
Collapse
Affiliation(s)
- Jie Chen
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Long Zhao
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu, Anhui, China
| | - Meng-Fan Xu
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Di Huang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Xiao-Long Sun
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Yu-Xin Zhang
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu, Anhui, China
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, China
| | - Hong-Mei Li
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu, Anhui, China
| | - Cheng-Zhu Wu
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu, Anhui, China
| |
Collapse
|
16
|
Lian H, Wang J, Yan S, Chen K, Jin L. An integrative analysis based on multiple cell death patterns identifies an immunosuppressive subtype and establishes a prognostic signature in lower-grade glioma. Ann Med 2024; 56:2412831. [PMID: 39387560 PMCID: PMC11469432 DOI: 10.1080/07853890.2024.2412831] [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: 11/30/2023] [Revised: 03/19/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Cell death modulates the biological behaviors of tumors. However, the comprehensive role of the multiple forms of cell death in lower-grade glioma (LGG) is unknown. METHODS We collected the transcriptional data of LGG patients from public repositories to comprehensively examine six cell death patterns (autophagy, apoptosis, cuproptosis, necroptosis, ferroptosis, and pyroptosis) in LGG samples and systematically correlated these patterns with patient survival, underlying biological processes, and drug sensitivity using serial bioinformatics analysis, clinical sample validation and in vitro assays. RESULTS We identified and independently validated three reproducible cell death-based clusters associated with distinct clinical outcomes and tumor microenvironment characteristics. The Tumor Immune Dysfunction and Exclusion algorithm was applied for predicting how these three clusters would respond to immune checkpoint blockade (ICB) therapy; we found potential resistance of cluster B to ICB therapy. We also performed drug screening to identify cluster-specific drugs. Furthermore, a scoring system, designated as the CDPM score, was developed to estimate the cell death patterns of patients with LGG; this system could predict both LGG patients' prognosis and immunotherapy efficacy. By performing multiplex immunofluorescence staining, we validated the correlations of GNAL expression with the molecular and clinical features of LGG in an independent LGG cohort. Finally, in vitro assays showed that GNAL promoted apoptosis and inhibited the proliferation of LGG cells. CONCLUSION The new cell death-based subtype system indicates several features of LGG biology and reveals novel insights into the use of precision medicine for treating LGG. The CDPM score could be used to predict the immunotherapy response and prognosis of LGG patients; moreover, it could indicate a novel direction for improving LGG management.
Collapse
Affiliation(s)
- Hao Lian
- Department of Traditional Chinese Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajia Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan Yan
- Pudong New District, Huamu Community Health Service Center, Shanghai, P.R. China
| | - Kui Chen
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lilun Jin
- Department of Traditional Chinese Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Tkachenko A, Havranek O. Erythronecroptosis: an overview of necroptosis or programmed necrosis in red blood cells. Mol Cell Biochem 2024; 479:3273-3291. [PMID: 38427167 DOI: 10.1007/s11010-024-04948-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/20/2024] [Indexed: 03/02/2024]
Abstract
Necroptosis is considered a programmed necrosis that requires receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3), and pore-forming mixed lineage kinase domain-like protein (MLKL) to trigger a regulated cell membrane lysis. Membrane rupture in necroptosis has been shown to fuel innate immune response due to release of damage-associated molecular patterns (DAMPs). Recently published studies indicate that mature erythrocytes can undergo necroptosis as well. In this review, we provide an outline of multiple cell death modes occurring in erythrocytes, discuss possible immunological aspects of diverse erythrocyte cell deaths, summarize available evidence related to the ability of erythrocytes to undergo necroptosis, outline key involved molecular mechanisms, and discuss the potential implication of erythrocyte necroptosis in the physiology and pathophysiology. Furthermore, we aim to highlight the interplay between necroptosis and eryptosis signaling in erythrocytes, emphasizing specific characteristics of these pathways distinct from their counterparts in nucleated cells. Thus, our review provides a comprehensive summary of the current knowledge of necroptosis in erythrocytes. To reflect critical differences between necroptosis of nucleated cells and necroptosis of erythrocytes, we suggest a term erythronecroptosis for necroptosis of enucleated cells.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 25250, Vestec, Czech Republic.
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 25250, Vestec, Czech Republic
- First Department of Internal Medicine-Hematology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
18
|
Dong ZK, Wang YF, Li WP, Jin WL. Neurobiology of cancer: Adrenergic signaling and drug repurposing. Pharmacol Ther 2024; 264:108750. [PMID: 39527999 DOI: 10.1016/j.pharmthera.2024.108750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Cancer neuroscience, as an emerging converging discipline, provides us with new perspectives on the interactions between the nervous system and cancer progression. As the sympathetic nervous system, in particular adrenergic signaling, plays an important role in the regulation of tumor activity at every hierarchical level of life, from the tumor cell to the tumor microenvironment, and to the tumor macroenvironment, it is highly desirable to dissect its effects. Considering the far-reaching implications of drug repurposing for antitumor drug development, such a large number of adrenergic receptor antagonists on the market has great potential as one of the means of antitumor therapy, either as primary or adjuvant therapy. Therefore, this review aims to summarize the impact of adrenergic signaling on cancer development and to assess the status and prospects of intervening in adrenergic signaling as a therapeutic tool against tumors.
Collapse
Affiliation(s)
- Zi-Kai Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Yong-Fei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Wei-Ping Li
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Urology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Wei-Lin Jin
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
19
|
Chen Q, Zhang C, Meng T, Yang K, Hu Q, Tong Z, Wang X. Prediction of clinical prognosis and drug sensitivity in hepatocellular carcinoma through the combination of multiple cell death pathways. Cell Biol Int 2024; 48:1816-1835. [PMID: 39192561 DOI: 10.1002/cbin.12235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/29/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024]
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignant tumor, highlighting a significant need for reliable predictive models to assess clinical prognosis, disease progression, and drug sensitivity. Recent studies have highlighted the critical role of various programmed cell death pathways, including apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, entotic cell death, NETotic cell death, parthanatos, lysosome-dependent cell death, autophagy-dependent cell death, alkaliptosis, oxeiptosis, and disulfidptosis, in tumor development. Therefore, by investigating these pathways, we aimed to develop a predictive model for HCC prognosis and drug sensitivity. We analyzed transcriptome, single-cell transcriptome, genomic, and clinical information using data from the TCGA-LIHC, GSE14520, GSE45436, and GSE166635 datasets. Machine learning algorithms were used to establish a cell death index (CDI) with seven gene signatures, which was validated across three independent datasets, showing that high CDI correlates with poorer prognosis. Unsupervised clustering revealed three molecular subtypes of HCC with distinct biological processes. Furthermore, a nomogram integrating CDI and clinical information demonstrated good predictive performance. CDI was associated with immune checkpoint genes and tumor microenvironment components using single-cell transcriptome analysis. Drug sensitivity analysis indicated that patients with high CDI may be resistant to oxaliplatin and cisplatin but sensitive to axitinib and sorafenib. In summary, our model offers a precise prediction of clinical outcomes and drug sensitivity for patients with HCC, providing valuable insights for personalized treatment strategies.
Collapse
Affiliation(s)
- QingKun Chen
- Department of Graduate School, Bengbu Medical University, Bengbu, China
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - ChenGuang Zhang
- Department of Graduate School, Bengbu Medical University, Bengbu, China
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - Tao Meng
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - Ke Yang
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - QiLi Hu
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - Zhong Tong
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| | - XiaoGang Wang
- Department of Hepatobiliary Surgery, The First People's Hospital of Hefei, Hefei, China
| |
Collapse
|
20
|
Bai F, Deng Y, Li L, Lv M, Razzokov J, Xu Q, Xu Z, Chen Z, Chen G, Chen Z. Advancements and challenges in brain cancer therapeutics. EXPLORATION (BEIJING, CHINA) 2024; 4:20230177. [PMID: 39713205 PMCID: PMC11655316 DOI: 10.1002/exp.20230177] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/02/2024] [Indexed: 12/24/2024]
Abstract
Treating brain tumors requires a nuanced understanding of the brain, a vital and delicate organ. Location, size, tumor type, and surrounding tissue health are crucial in developing treatment plans. This review comprehensively summarizes various treatment options that are available or could be potentially available for brain tumors, including physical therapies (radiotherapy, ablation therapy, photodynamic therapy, tumor-treating field therapy, and cold atmospheric plasma therapy) and non-physical therapies (surgical resection, chemotherapy, targeted therapy, and immunotherapy). Mechanisms of action, potential side effects, indications, and latest developments, as well as their limitations, are highlighted. Furthermore, the requirements for personalized, multi-modal treatment approaches in this rapidly evolving field are discussed, emphasizing the balance between efficacy and patient safety.
Collapse
Affiliation(s)
- Fan Bai
- Paul C Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health EngineeringShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- Advanced Therapeutic CenterNational Innovation Center for Advanced Medical DevicesShenzhenChina
| | - Yueyang Deng
- Department of Biomedical EngineeringMcGill UniversityMontrealQuebecCanada
- Rosalind & Morris Goodman Cancer InstituteMcGill UniversityMontrealQuebecCanada
| | - Long Li
- Paul C Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health EngineeringShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesShenzhenGuangdongP. R. China
| | - Ming Lv
- Department of Medical EngineeringMedical Supplies Center of Chinese PLA General HospitalBeijingChina
| | - Jamoliddin Razzokov
- Institute of Fundamental and Applied ResearchNational Research University TIIAMETashkentUzbekistan
- Laboratory of Experimental BiophysicsCentre for Advanced TechnologiesTashkentUzbekistan
- Department of Biomedical EngineeringTashkent State Technical UniversityTashkentUzbekistan
| | - Qingnan Xu
- Paul C Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health EngineeringShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Zhen Xu
- Paul C Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health EngineeringShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Zhaowei Chen
- Institute of Food Safety and Environment MonitoringMOE Key Laboratory for Analytical Science of Food Safety and BiologyCollege of ChemistryFuzhou UniversityFuzhouChina
| | - Guojun Chen
- Department of Biomedical EngineeringMcGill UniversityMontrealQuebecCanada
- Rosalind & Morris Goodman Cancer InstituteMcGill UniversityMontrealQuebecCanada
| | - Zhitong Chen
- Paul C Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health EngineeringShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- Advanced Therapeutic CenterNational Innovation Center for Advanced Medical DevicesShenzhenChina
- University of Chinese Academy of SciencesShenzhenGuangdongP. R. China
- Key Laboratory of Biomedical Imaging Science and SystemChinese Academy of SciencesShenzhenChina
| |
Collapse
|
21
|
Lotfi MS, Rassouli FB. Navigating the complexities of cell death: Insights into accidental and programmed cell death. Tissue Cell 2024; 91:102586. [PMID: 39426124 DOI: 10.1016/j.tice.2024.102586] [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: 08/21/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Cell death is a critical biological phenomenon that can be categorized into accidental cell death (ACD) and programmed cell death (PCD), each exhibiting distinct signaling, mechanistic and morphological characteristics. This paper provides a comprehensive overview of seven types of ACD, including coagulative, liquefactive, caseous, fat, fibrinoid, gangrenous and secondary necrosis, discussing their pathological implications in conditions such as ischemia and inflammation. Additionally, we review eighteen forms of PCD, encompassing autophagy, apoptosis, necroptosis, pyroptosis, paraptosis, ferroptosis, anoikis, entosis, NETosis, eryptosis, parthanatos, mitoptosis, and newly recognized types such as methuosis, autosis, alkaliptosis, oxeiptosis, cuprotosis and erebosis. The implications of these cell death modalities for cellular processes, development, and disease-particularly in the context of neoplastic and neurodegenerative disorders-are also covered. Furthermore, we explore the crosstalk between various forms of PCD, emphasizing how apoptotic mechanisms can influence pathways like necroptosis and pyroptosis. Understanding this interplay is crucial for elucidating cellular responses to stress, as well as for its potential relevance in clinical applications and therapeutic strategies. Future research should focus on clarifying the molecular mechanisms that govern different forms of PCD and their interactions.
Collapse
Affiliation(s)
- Mohammad-Sadegh Lotfi
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fatemeh B Rassouli
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
22
|
Mao S, Zhao Y, Xiong H, Gong C. Excavating regulated cell death signatures to predict prognosis, tumor microenvironment and therapeutic response in HR+/HER2- breast cancer. Transl Oncol 2024; 50:102117. [PMID: 39241556 PMCID: PMC11406102 DOI: 10.1016/j.tranon.2024.102117] [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: 06/04/2024] [Revised: 07/25/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024] Open
Abstract
Regulated cell death (RCD) has been documented to have great potentials for discovering novel biomarkers and therapeutic targets in malignancies. But its role and clinical value in HR+/HER2- breast cancer, the most common subtype of breast cancer, are obscure. In this study, we comprehensively explored 12 types of RCD patterns and found extensive mutations and dysregulations of RCD genes in HR+/HER2- breast cancer. A prognostic RCD scoring system (CDScore) based on six critical genes (LEF1, SLC7A11, SFRP1, IGFBP6, CXCL2, STXBP1) was constructed, in which a high CDScore predicts poor prognosis. The expressions and prognostic value of LEF1 and SFRP1were also validated in our tissue microarrays. The nomogram established basing on CDScore, age and TNM stage performed satisfactory in predicting overall survival, with an area under the ROC curve of 0.89, 0.82 and 0.8 in predicting 1-year, 3-year and 5-year overall survival rates, respectively. Furthermore, CDScore was identified to be correlated with tumor microenvironments and immune checkpoints by excavation of bulk and single-cell sequencing data. Patients in CDScore high group might be resistant to standard chemotherapy and target therapy. Our results underlined the potential effects and importance of RCD in HR+/HER2- breast cancer and provided novel biomarkers and therapeutic targets for HR+/HER2- breast cancer patients.
Collapse
Affiliation(s)
- Shuangshuang Mao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yuanyuan Zhao
- Department of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
23
|
Liao Y, Zhang W, Zhou M, Zhu C, Zou Z. Ubiquitination in pyroptosis pathway: A potential therapeutic target for sepsis. Cytokine Growth Factor Rev 2024; 80:72-86. [PMID: 39294049 DOI: 10.1016/j.cytogfr.2024.09.001] [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: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Sepsis remains a significant clinical challenge, causing numerous deaths annually and representing a major global health burden. Pyroptosis, a unique form of programmed cell death characterized by cell lysis and the release of inflammatory mediators, is a crucial factor in the pathogenesis and progression of sepsis, septic shock, and organ dysfunction. Ubiquitination, a key post-translational modification influencing protein fate, has emerged as a promising target for managing various inflammatory conditions, including sepsis. This review integrates the current knowledge on sepsis, pyroptosis, and the ubiquitin system, focusing on the molecular mechanisms of ubiquitination within pyroptotic pathways activated during sepsis. By exploring how modulating ubiquitination can regulate pyroptosis and its associated inflammatory signaling pathways, this review provides insights into potential therapeutic strategies for sepsis, highlighting the need for further research into these complex molecular networks.
Collapse
Affiliation(s)
- Yan Liao
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Wangzheqi Zhang
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Miao Zhou
- Department of Anesthesiology, the Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Chenglong Zhu
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| | - Zui Zou
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
24
|
Yan K, Zhang W, Song H, Xu X. Sphingolipid metabolism and regulated cell death in malignant melanoma. Apoptosis 2024; 29:1860-1878. [PMID: 39068623 DOI: 10.1007/s10495-024-02002-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] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Malignant melanoma (MM) is a highly invasive and therapeutically resistant skin malignancy, posing a significant clinical challenge in its treatment. Programmed cell death plays a crucial role in the occurrence and progression of MM. Sphingolipids (SP), as a class of bioactive lipids, may be associated with many kinds of diseases. SPs regulate various forms of programmed cell death in tumors, including apoptosis, necroptosis, ferroptosis, and more. This review will delve into the mechanisms by which different types of SPs modulate various forms of programmed cell death in MM, such as their regulation of cell membrane permeability and signaling pathways, and how they influence the survival and death fate of MM cells. An in-depth exploration of the role of SPs in programmed cell death in MM aids in unraveling the molecular mechanisms of melanoma development and holds significant importance in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Kexin Yan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, China
| | - Wei Zhang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, China
| | - Hao Song
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, China.
| | - Xiulian Xu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, China.
| |
Collapse
|
25
|
Wang H, Wang T, Yan S, Tang J, Zhang Y, Wang L, Xu H, Tu C. Crosstalk of pyroptosis and cytokine in the tumor microenvironment: from mechanisms to clinical implication. Mol Cancer 2024; 23:268. [PMID: 39614288 PMCID: PMC11607834 DOI: 10.1186/s12943-024-02183-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
In the realm of cancer research, the tumor microenvironment (TME) plays a crucial role in tumor initiation and progression, shaped by complex interactions between cancer cells and surrounding non-cancerous cells. Cytokines, as essential immunomodulatory agents, are secreted by various cellular constituents within the TME, including immune cells, cancer-associated fibroblasts, and cancer cells themselves. These cytokines facilitate intricate communication networks that significantly influence tumor initiation, progression, metastasis, and immune suppression. Pyroptosis contributes to TME remodeling by promoting the release of pro-inflammatory cytokines and sustaining chronic inflammation, impacting processes such as immune escape and angiogenesis. However, challenges remain due to the complex interplay among cytokines, pyroptosis, and the TME, along with the dual effects of pyroptosis on cancer progression and therapy-related complications like cytokine release syndrome. Unraveling these complexities could facilitate strategies that balance inflammatory responses while minimizing tissue damage during therapy. This review delves into the complex crosstalk between cytokines, pyroptosis, and the TME, elucidating their contribution to tumor progression and metastasis. By synthesizing emerging therapeutic targets and innovative technologies concerning TME, this review aims to provide novel insights that could enhance treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Tao Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Shuxiang Yan
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jinxin Tang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yibo Zhang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Liming Wang
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410011, China.
| | - Haodong Xu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Shenzhen Research Institute of Central South University, Guangdong, 518063, China.
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
26
|
Wan J, Zhao J, Fang X. Dynamics of the immune microenvironment and immune cell PANoptosis in colorectal cancer: recent advances and insights. Front Immunol 2024; 15:1502257. [PMID: 39676861 PMCID: PMC11638180 DOI: 10.3389/fimmu.2024.1502257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most significant oncological threats to human health globally. Patients often exhibit a high propensity for tumor recurrence and metastasis post-surgery, resulting in suboptimal prognoses. One of the underlying reasons for the metastatic potential of CRC is the sustained abnormal state of the tumor immune microenvironment, particularly characterized by the atypical death of critical immune cells. In recent years, a novel concept of cell death known as PANoptosis has emerged. This form of cell death is regulated by the PANoptosome complex and encompasses key features of apoptosis, pyroptosis, and necroptosis, yet cannot be entirely substituted by any of these processes alone. Due to its widespread occurrence and complex mechanisms, PANoptosis has been increasingly reported in various malignancies, enhancing our understanding of its pathological mechanisms, particularly in the context of CRC. However, the characteristics of immune cell PANoptosis within the CRC immune microenvironment have not been thoroughly elucidated. In this review, we focus on the impact of CRC progression on various immune cell types and summarize the distinctive features of immune cell PANoptosis. Furthermore, we highlight the future research trends and challenges associated with the mechanisms of immune cell PANoptosis in CRC.
Collapse
Affiliation(s)
- Jinlong Wan
- Department of Gastroenterology, Gaozhou People's Hospital, Maoming, China
| | - Jianzhong Zhao
- Department of Clinical Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Xiaolu Fang
- Department of Clinical Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| |
Collapse
|
27
|
Barakat R, Chatterjee J, Mu R, Qi X, Gu X, Smirnov I, Cobb O, Gao K, Barnes A, Kipnis J, Gutmann DH. Human single cell RNA-sequencing reveals a targetable CD8 + exhausted T cell population that maintains mouse low-grade glioma growth. Nat Commun 2024; 15:10312. [PMID: 39609412 PMCID: PMC11605098 DOI: 10.1038/s41467-024-54569-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
In solid cancers, T cells typically function as cytotoxic effectors to limit tumor growth, prompting therapies that capitalize upon this antineoplastic property (immune checkpoint inhibition; ICI). Unfortunately, ICI treatments have been largely ineffective for high-grade brain tumors (gliomas; HGGs). Leveraging several single-cell RNA sequencing datasets, we report greater CD8+ exhausted T cells in human pediatric low-grade gliomas (LGGs) relative to adult and pediatric HGGs. Using several preclinical mouse LGG models (Nf1-OPG mice), we show that these PD1+/TIGIT+ CD8+ exhausted T cells are restricted to the tumor tissue, where they express paracrine factors necessary for OPG growth. Importantly, ICI treatments with α-PD1 and α-TIGIT antibodies attenuate Nf1-OPG tumor proliferation through suppression of two cytokine (Ccl4 and TGFβ)-mediated mechanisms, rather than by T cell-mediated cytotoxicity, as well as suppress monocyte-controlled T cell chemotaxis. Collectively, these findings establish a previously unrecognized function for CD8+ exhausted T cells as specialized regulators of LGG maintenance.
Collapse
Affiliation(s)
- Rasha Barakat
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rui Mu
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xuanhe Qi
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xingxing Gu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Igor Smirnov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Olivia Cobb
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Karen Gao
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Angelica Barnes
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
28
|
Zhuang X, Chen P, Yang K, Yang R, Man X, Wang R, Shi Y. MT1E in AML: a gateway to understanding regulatory cell death and immunotherapeutic responses. J Leukoc Biol 2024; 116:1515-1529. [PMID: 38943611 PMCID: PMC11599122 DOI: 10.1093/jleuko/qiae151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 06/04/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024] Open
Abstract
Regulated cell death (RCD) plays a crucial role in the initiation and progression of tumors, particularly in acute myeloid leukemia (AML). This study investigates the prognostic importance of RCD-related genes in AML and their correlation with immune infiltration. We combined TCGA and GTEx data, analyzing 1,488 RCD-related genes, to develop a predictive model using LASSO regression and survival analysis. The model's accuracy was validated against multiple databases, examining immune cell infiltration, therapy responses, and drug sensitivity among risk groups. RT-qPCR confirmed MT1E expression in AML patients and healthy bone marrow. CCK8 and Transwell assays measured cell proliferation, adhesion, migration, and invasion, while flow cytometry and Western blotting assessed apoptosis and protein expression. We developed a prognostic model using 10 RCD methods, which demonstrated strong predictive ability, showing an inverse correlation between age and risk scores with survival in AML patients. Functional enrichment analysis of the model is linked to immune modulation pathways. RT-qPCR revealed significantly lower MT1E expression in AML vs healthy bone marrow (P < 0.05). Consequently, experiments were designed to assess the function of MT1E overexpression. Findings indicated that MT1E overexpression showed it significantly reduced THP-1 cell proliferation and adhesion (P < 0.001), decreased migration (P < 0.001), and invasiveness (P < 0.05), and increased apoptosis (P < 0.05), with a notable rise in Caspase3 expression. A novel AML RCD risk model was developed, showing promise as a prognostic marker for evaluating outcomes and immune therapy effectiveness. Insights into MT1E's impact on AML cell proliferation and apoptosis open possibilities for improving patient outcomes and devising personalized treatment strategies.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Immunotherapy
- Apoptosis
- Cell Proliferation
- Prognosis
- Male
- Female
- Cell Line, Tumor
- Cell Movement
- Middle Aged
- Gene Expression Regulation, Leukemic
Collapse
Affiliation(s)
- Xin Zhuang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Peng Chen
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Kaiqian Yang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Rong Yang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Xiaoying Man
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Ruochen Wang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Yifen Shi
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
- Qinghai Province Women and Children's Hospital, Xining, Qinghai Province 810000, China
| |
Collapse
|
29
|
Zheng Y, Sun J, Luo Z, Li Y, Huang Y. Emerging mechanisms of lipid peroxidation in regulated cell death and its physiological implications. Cell Death Dis 2024; 15:859. [PMID: 39587094 PMCID: PMC11589755 DOI: 10.1038/s41419-024-07244-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: 08/09/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Abstract
Regulated cell death (RCD) refers to the form of cell death that can be regulated by various biomacromolecules. Each cell death modalities have their distinct morphological changes and molecular mechanisms. However, intense evidences suggest that lipid peroxidation can be the common feature that initiates and propagates the cell death. Excessive lipid peroxidation alters the property of membrane and further damage the proteins and nucleic acids, which is implicated in various human pathologies. Here, we firstly review the classical chain process of lipid peroxidation, and further clarify the current understanding of the myriad roles and molecular mechanisms of lipid peroxidation in various RCD types. We also discuss how lipid peroxidation involves in diseases and how such intimate association between lipid peroxidation-driven cell death and diseases can be leveraged to develop rational therapeutic strategies.
Collapse
Affiliation(s)
- Yongxin Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
| | - Junlu Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Zhiting Luo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Yimin Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China.
- State Key Laboratory of Respiratory Diseases, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Yongbo Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China.
- State Key Laboratory of Respiratory Diseases, Guangzhou, China.
| |
Collapse
|
30
|
Pavlović S, Petrović B, Ćoćić D, Schreurer A, Sretenović S, Nešić MD, Nišavić M, Maric Z, Stanisavljević I, Ćorović I, Simović Marković B, Maric V, Jovanović I, Radić G, Radisavljević S, Jovanović Stević S. New Pd(II)-pincer type complexes as potential antitumor drugs: synthesis, nucleophilic substitution reactions, DNA/HSA interaction, molecular docking study and cytotoxic activity. Dalton Trans 2024; 53:18560-18574. [PMID: 39470017 DOI: 10.1039/d4dt02549k] [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: 10/30/2024]
Abstract
Two new complexes of Pd(II), [Pd(L1)Cl]Cl (Pd1) and [Pd(L2)Cl]Cl (Pd2), (where L1 = N2,N6-bis(5-methylthiazol-2-yl)pyridine-2,6-dicarboxamide and L2 = N2,N6-di(benzo[d]thiazol-2-yl)pyridine-2.6-dicarboxamide) were synthesized. Characterization of the complexes was performed using elemental analysis, IR, 1H NMR spectroscopy and MALDI-TOF mass spectrometry. The nucleophilic substitution reactions of complexes with L-Methionine (L-Met), L-Cysteine (L-Cys) and guanosine-5'-monophosphate (5'-GMP) were studied by stopped-flow method at physiological conditions (pH = 7.2 and 37 °C). Complex Pd1 was more reactive than Pd2 in all studied reactions, while the order of reactivity of the selected ligands was: L-Met > L-Cys > 5'-GMP. The interaction of complexes with calf thymus-DNA (CT-DNA) was studied by Uv-Vis absorption and fluorescence emission spectroscopy. Competitive binding studies with intercalative agent ethidium bromide (EB) and minor groove binder Hoechst 33258 were performed as well. Both complexes interacted with DNA through intercalation and minor groove binding, where the latter was preferred. Additionally, the interaction of Pd1 and Pd2 complexes with human serum albumin (HSA) was studied employing fluorescence quenching spectroscopy. The results indicate a moderate binding affinity of complexes, with slightly stronger binding of the Pd1. Fluorescence competition experiments with site-markers (eosin Y and ibuprofen) for HSA were used to locate the binding site of Pd1 to the HSA. Additionally, the interaction with DNA and HSA was studied by molecular docking and the revealed results were in good agreement with the experimentally obtained ones. Pd1 complex exhibited cytotoxicity toward human (HCT116) and mouse cell lines (CT26) of colorectal cancer, mouse (4T1) and human (MDA-MB468) breast cancer lines and non-cancerous mouse mesenchymal stem cells (mMSC). In addition, Pd1 complex demonstrated significant selectivity towards cancer cells over non-cancerous mMSC, indicating a high potential to eliminate malignant cells without affecting normal cells. It induced apoptosis in CT26 cells, effectively arrested the cell cycle in the S phase, and selectively down-regulated cyclin D and cyclin E. Moreover, it can alter the expression of cell cycle regulators by increasing p21 and decreasing p-AKT. These findings confirm its ability to disrupt key tumor cell survival signals and suggest that the Pd1 complex is a potent candidate for effective cancer treatment.
Collapse
Affiliation(s)
- Sladjana Pavlović
- University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Biljana Petrović
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Dušan Ćoćić
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Andreas Schreurer
- Inorganic Chemistry, Department of Chemistry and Pharmacy, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Snežana Sretenović
- University of Kragujevac, Faculty of Medicinal Science, Department of Internal Medicine, Kragujevac, Serbia
| | - Maja D Nešić
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000, Serbia
| | - Marija Nišavić
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000, Serbia
| | - Zorana Maric
- University of East Sarajevo, Faculty of Medicine, Studentska 5, 73300 Foca, BiH
| | - Isidora Stanisavljević
- University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Irfan Ćorović
- General Hospital of Novi Pazar, Department of Internal Medicine, Generala Živkovića 1, 36300 Novi Pazar, Serbia
| | - Bojana Simović Marković
- University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Veljko Maric
- University of East Sarajevo, Faculty of Medicine, Studentska 5, 73300 Foca, BiH
| | - Ivan Jovanović
- University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Gordana Radić
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, 34000 Kragujevac, Serbia.
| | - Snežana Radisavljević
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Snežana Jovanović Stević
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, 34000 Kragujevac, Serbia.
| |
Collapse
|
31
|
Cao Z, Tian K, Ran Y, Zhou H, Zhou L, Ding Y, Tang X. Beclin-1: a therapeutic target at the intersection of autophagy, immunotherapy, and cancer treatment. Front Immunol 2024; 15:1506426. [PMID: 39650649 PMCID: PMC11621085 DOI: 10.3389/fimmu.2024.1506426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/01/2024] [Indexed: 12/11/2024] Open
Abstract
The significant identification of Beclin-1's function in regulating autophagy flow signified a significant progression in our understanding of cellular operations. Beclin-1 acts as a scaffold for forming the PI3KC3 complex, controlling autophagy and cellular trafficking processes in a complicated way. This intricate protein has garnered considerable attention due to its substantial impact on the development of tumors. Strong evidence indicates Beclin-1 plays a critical role in controlling autophagy in various human cancer types and its intricate connection with apoptosis and ferroptosis. The potential of Beclin-1 as a viable target for cancer therapy is highlighted by its associations with key autophagy regulators such as AMPK, mTOR, and ATGs. Beclin-1 controls the growth and dissemination of tumors by autophagy. It also affects how tumors react to therapies such as chemotherapy and radiation therapy. The role of Beclin-1 in autophagy can influence apoptosis, depending on whether it supports cell survival or leads to cell death. Beclin-1 plays a crucial role in ferroptosis by increasing ATG5 levels, which in turn promotes autophagy-triggered ferroptosis. Finally, we analyzed the possible function of Beclin-1 in tumor immunology and drug sensitivity in cancers. In general, Beclin-1 has a significant impact on regulating autophagy, offering various potentials for medical intervention and altering our understanding of cancer biology.
Collapse
Affiliation(s)
- Zhumin Cao
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Ke Tian
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Yincheng Ran
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Haonan Zhou
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Lei Zhou
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Yana Ding
- Department of Hepatobiliary Surgery, District Traditional Chinese Medicine Hospital, Chongqing, China
| | - Xiaowei Tang
- Department of Hepatobiliary Surgery, District Traditional Chinese Medicine Hospital, Chongqing, China
| |
Collapse
|
32
|
Tang N, Chen Y, Su Y, Zhang S, Huang T. The role of disulfidptosis-associated LncRNA-LINC01137 in Osteosarcoma Biology and its regulatory effects on macrophage polarization. Funct Integr Genomics 2024; 24:219. [PMID: 39576417 DOI: 10.1007/s10142-024-01504-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: 07/02/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 11/24/2024]
Abstract
The objective of this research is to investigate the function of long non-coding RNA (lncRNA) associated with disulfidptosis, particularly LINC01137, in osteosarcoma (OS), and its impact on macrophage polarization and the tumor immune microenvironment (TME), with the goal of identifying new prognostic biomarkers and therapeutic targets. Utilizing the OS transcriptome dataset from the TARGET database, differentially expressed lncRNAs related to disulfidptosis were identified. The functional mechanisms of LINC01137, which affect cell proliferation, migration, invasiveness, programmed cell death, and macrophage orientation, were explored using the full suite of analyses provided by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), alongside a diverse array of laboratory experiments, including an in vivo osteosarcoma xenograft model in BALB/c nude mice to assess the impact of LINC01137 knockdown on tumor growth. Among three lncRNAs identified that were distinctly linked to disulfidptosis, LINC01137 showed a notable increase in expression within OS cell lines. Silencing LINC01137 led to a marked decrease in the abilities of cell proliferation, migration, and invasiveness, simultaneously enhancing programmed cell death and facilitating the process of epithelial-mesenchymal transition (EMT). In vivo experiments further confirmed that LINC01137 knockdown significantly suppressed tumor growth in osteosarcoma xenograft models, aligning with the in vitro findings. Associated with disulfidptosis, LINC01137 is pivotal in osteosarcoma development through its enhancement of tumor cell proliferation, migration, and invasiveness, as well as its modification of macrophage orientation within the TME. Given its significance, LINC01137 merits exploration as a prognostic indicator, necessitating detailed studies on its regulatory functions and potential in therapy.
Collapse
Affiliation(s)
- Ning Tang
- Orthopaedic Department, The Second Xiangya Hospital of Central South Unniversity, Hunan Province, Changsha, China
- Orthopaedic Department, The Third Xiangya Hospital of Central South University, Hunan Province, Changsha, China
| | - Yifan Chen
- Orthopaedic Department, The Second Xiangya Hospital of Central South Unniversity, Hunan Province, Changsha, China
| | - Yang Su
- Orthopaedic Department, The Second Xiangya Hospital of Central South Unniversity, Hunan Province, Changsha, China
| | - Shengqun Zhang
- Orthopaedic Department, The Second Xiangya Hospital of Central South Unniversity, Hunan Province, Changsha, China
| | - Tianlong Huang
- Orthopaedic Department, The Second Xiangya Hospital of Central South Unniversity, Hunan Province, Changsha, China.
| |
Collapse
|
33
|
Chen T, Liang K, Wang J, Li J, Xue X, Hao Y, Liang H, Ren H, Xiao H, Ge J, Tang B. An Aged Tree with a New Bloom: A Simple Spatiotemporal Programming Strategy Enables Carbon Dot Photosensitizers to Regulate Cell Pyroptosis for Enhanced Tumor Photodynamic-Immunotherapy. NANO LETTERS 2024; 24:14709-14719. [PMID: 39504147 DOI: 10.1021/acs.nanolett.4c03913] [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: 11/08/2024]
Abstract
Pyroptosis induced by photodynamic therapy (PDT) is a promising field in both PDT and immunotherapy for tumors. However, effectively inducing tumor cell pyroptosis while triggering a strong immune response using current photosensitizers remains challenging. Herein, the developed positively charged carbon dots (PCDs) nanoPSs were utilized to modulate tumor cell pyroptosis for the first time through a simple spatiotemporal programming strategy. Briefly, PCDs enabled precisely time-dependent targeting of the cell membrane or lysosome. Upon light irradiation, in vitro studies revealed that lysosome-targeted PDT primarily induced apoptosis, while membrane-targeted PDT triggered pyroptosis, resulting in enhanced PDT efficacy and robust activation of the immune response. Conclusively, in vivo studies demonstrated that PCDs could serve as a novel pyroptosis nanotuner for enhanced photodynamic-immunotherapy, thereby simultaneously eliminating primary tumors and inhibiting distant tumor growth and metastases. This spatiotemporal programming strategy unprecedentedly offers a rejuvenation of aged PSs and expands the biomedical use of CDs in immunotherapy.
Collapse
Affiliation(s)
- Tiejin Chen
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ke Liang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jian Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jian Li
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiaokuang Xue
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yongliang Hao
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Huanyi Liang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Haohui Ren
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Jiechao Ge
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
- Laoshan Laboratory, Qingdao, 266237, P. R. China
| |
Collapse
|
34
|
Hou G, Chen Y, Lei H, Lu S, Cheng L. Nanomaterials-Induced PANoptosis: A Promising Anti-Tumor Strategy. Angew Chem Int Ed Engl 2024:e202419649. [PMID: 39560000 DOI: 10.1002/anie.202419649] [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: 10/11/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/20/2024]
Abstract
Malignant tumors pose a significant threat to global public health. Promoting programmed cell death in cancer cells has become a critical strategy for cancer treatment. PANoptosis, a newly discovered form of regulated cell death, integrates key molecular components of pyroptosis, apoptosis, and necroptosis, activating these three death pathways simultaneously to achieve synergistic multi-mechanistic killing. PANoptosis significantly inhibits cancer cell growth and resistance and activates strong anti-tumor immune response, making tumor-specific induction of PANoptosis a potential cancer therapeutic strategy. Currently, cancer treatment research related to PANoptosis is focused mainly on the development of small molecules and cytokines. However, these approaches still face limitations in terms of metabolic stability and tumor specificity. The unique physicochemical properties and biological activities of nanomaterials hold significant promise for optimizing PANoptosis induction strategies. This review summarizes the concept and mechanisms of PANoptosis, highlights the latest applications of nanoagents in PANoptosis-based anti-cancer therapy, and discusses the challenges and future directions for clinical translation. It is hoped that this review will inspire further exploration and development of PANoptosis-based cancer treatments, providing new perspectives for researchers in the field.
Collapse
Affiliation(s)
- Guanghui Hou
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Youdong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
35
|
Guo F, Ji X, Xiong C, Sun H, Liang Z, Yan-Do R, Gai B, Gao F, Huang L, Li Z, Kuang BY, Shi P. Single-cell encoded gene silencing for high-throughput combinatorial siRNA screening. Nat Commun 2024; 15:9985. [PMID: 39562763 DOI: 10.1038/s41467-024-53419-7] [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: 03/14/2024] [Accepted: 10/09/2024] [Indexed: 11/21/2024] Open
Abstract
The use of combinatorial siRNAs shows great promise for drug discovery, but the identification of safe and effective siRNA combinations remains challenging. Here, we develop a massively multiplexed technology for systematic screening of siRNA-based cocktail therapeutics. We employ composite micro-carriers that are responsive to near infrared light and magnetic field to achieve photoporation-facilitated siRNA transfection to individual cells. Thus, randomized gene silencing by different siRNA formulations can be performed with high-throughput single-cell-based analyses. For screening anti-cancer siRNA cocktails, we test more than 1300 siRNA combinations for knocking down multiple genes related to tumor growth, discovering effective 3-siRNA formulations with an emphasis on the critical role of inhibiting Cyclin D1 and survivin, along with their complementary targets for synergic efficacy. This approach enables orders of magnitude reduction in time and cost associated with largescale siRNA screening, and resolves key insights to siRNA pharmacology that are not permissive to existing methods.
Collapse
Affiliation(s)
- Feng Guo
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, 999077, China
| | - Xianglin Ji
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Chuxiao Xiong
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Hailiang Sun
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Zhenghua Liang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, 999077, China
| | - Richard Yan-Do
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, 999077, China
| | - Baowen Gai
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Feng Gao
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Linfeng Huang
- Wang-Cai Biochemistry Lab, Division of Natural and Applied Sciences & Global Health Research Center, Duke Kunshan University, Kunshan, Jiangsu, China
| | - Zhongping Li
- Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Becki Yi Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, 999077, China
| | - Peng Shi
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China.
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, 999077, China.
- Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China.
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, 518000, China.
| |
Collapse
|
36
|
Wang T, Liu Y, Kong J, Liu J. Identification of a novel molecular classification for hepatocellular carcinoma based on disulfideptosis-related genes and its potential prognostic significance. J Cancer Res Clin Oncol 2024; 150:506. [PMID: 39551857 PMCID: PMC11570565 DOI: 10.1007/s00432-024-06031-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/09/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Globally, hepatocellular carcinoma (HCC) is one of the most prevalent and deadly malignant tumors. A recent study proposed disulfidptosis, a novel form of regulated cell death (RCD), offering a new avenue for identifying tumor prognosis biomarkers and developing novel therapeutic targets. METHODS Based on the expression data of 14 disulfideptosis-related genes extracted from public databases, a new molecular classification of HCC called the "disulfidptosis score" was constructed and its relationship to tumor immunity and prognosis was evaluated. RESULTS Based on the expression of disulfideptosis-related genes, we performed cluster analysis on HCC samples from the TCGA cohort, which classified these patients into three clusters: A, B, and C, and the differentially expressed genes of different clusters were analyzed. A disulfidptosis score model was constructed by differentially expressed genes associated with prognosis. Univariate and multivariate COX regression analysis showed that disulfidptosis score was an independent prognostic factor for HCC. In addition, in various disulfidptosis score groups, notable disparities were observed concerning the tumor immune microenvironment as well as the expression of immune checkpoint. CONCLUSION Disulfidptosis score have an important role in predicting HCC prognosis and help guide us in providing better immunotherapy options for patients.
Collapse
Affiliation(s)
- Tao Wang
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Jinan, 250000, China
| | - Yong Liu
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Jinan, 250000, China
| | - Junjie Kong
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Jinan, 250000, China
| | - Jun Liu
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Jinan, 250000, China.
| |
Collapse
|
37
|
Mishra S, Shelar SB, Rout S, Hassan PA, Barick KC, Agarwal N. Enhanced Singlet Oxygen Generation in Aggregates of Naphthalene-Fused BODIPY and Its Application in Photodynamic Therapy. ACS APPLIED BIO MATERIALS 2024; 7:7207-7218. [PMID: 39445398 DOI: 10.1021/acsabm.4c00804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Several reports are available on aggregation-induced emission and its applications in biomedical imaging and other material sciences. However, enhancement of singlet oxygen generation in nanoaggregates is rarely reported. Here, we report the synthesis of Naph-BODIPY Br2, which absorbs at 661 nm (monomer) with a high molar absorption coefficient. The presence of bromine promotes intersystem crossing, thereby enhancing the singlet oxygen quantum yield (ΦΔ ∼ 0.50 in methanol). In order to increase hydrophilicity, we developed Naph-BODIPY Br2 nanoaggregates (∼100 nm), which demonstrated aggregation-induced properties and exhibited a bathochromic shift with an absorption maximum at 757 nm. The bathochromic shift in the UV-vis spectra due to aggregation is corroborated by TD-DFT analysis. The computational data also confirm the presence of a low-lying triplet state, which enhances the generation of singlet oxygen, making it effective for photodynamic therapy. These aggregates showed excellent singlet oxygen generation in aqueous media, compared to their monomeric form and standard methylene blue. Their hydrophilic nature and high singlet oxygen generation enabled significant phototoxicity against human carcinoma cells with IC50 values of 4.06 ± 0.01 and 4.09 ± 0.1 μM, respectively, for MCF-7 and A549 cells upon 5 min exposure to light. Moreover, their phototoxicity further increases with an increasing exposure time of light for both cell lines. Notably, Naph-BODIPY Br2 nanoaggregates exhibited nearly zero dark cell toxicity and effectively induced apoptosis in cancer cells upon light activation, highlighting their potential as powerful photosensitizers for photodynamic cancer therapy.
Collapse
Affiliation(s)
- Sneha Mishra
- School of Chemical Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Kalina, Santacruz (E), Mumbai 400098, India
| | | | - Saiprakash Rout
- School of Chemical Sciences, National Institute of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni, Khurda752050,India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Puthusserickal A Hassan
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - K C Barick
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Neeraj Agarwal
- School of Chemical Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Kalina, Santacruz (E), Mumbai 400098, India
| |
Collapse
|
38
|
Zhang X, Tang B, Luo J, Yang Y, Weng Q, Fang S, Zhao Z, Tu J, Chen M, Ji J. Cuproptosis, ferroptosis and PANoptosis in tumor immune microenvironment remodeling and immunotherapy: culprits or new hope. Mol Cancer 2024; 23:255. [PMID: 39543600 PMCID: PMC11566504 DOI: 10.1186/s12943-024-02130-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/19/2024] [Indexed: 11/17/2024] Open
Abstract
Normal life requires cell division to produce new cells, but cell death is necessary to maintain balance. Dysregulation of cell death can lead to the survival and proliferation of abnormal cells, promoting tumor development. Unlike apoptosis, necrosis, and autophagy, the newly recognized forms of regulated cell death (RCD) cuproptosis, ferroptosis, and PANoptosis provide novel therapeutic strategies for tumor treatment. Increasing research indicates that the death of tumor and immune cells mediated by these newly discovered forms of cell death can regulate the tumor microenvironment (TME) and influence the effectiveness of tumor immunotherapy. This review primarily elucidates the molecular mechanisms of cuproptosis, ferroptosis, and PANoptosis and their complex effects on tumor cells and the TME. This review also summarizes the exploration of nanoparticle applications in tumor therapy based on in vivo and in vitro evidence derived from the induction or inhibition of these new RCD pathways.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Bufu Tang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jinhua Luo
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Yang Yang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Qiaoyou Weng
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Shiji Fang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China
| | - Jianfei Tu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China.
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China.
| | - Jiansong Ji
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Csaenter of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
- Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
- School of Medcine, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000, China.
| |
Collapse
|
39
|
Choroba K, Zowiślok B, Kula S, Machura B, Maroń AM, Erfurt K, Marques C, Cordeiro S, Baptista PV, Fernandes AR. Optimization of Antiproliferative Properties of Triimine Copper(II) Complexes. J Med Chem 2024; 67:19475-19502. [PMID: 39496093 PMCID: PMC11571215 DOI: 10.1021/acs.jmedchem.4c01806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
Cu(II) complexes with 2,2':6',2″-terpyridines (terpy) and 2,6-bis(thiazol-2-yl)pyridines (dtpy) with 1- or 2-naphtyl and methoxy-naphtyl were synthesized to elucidate the impact of the triimine core, naphtyl linking mode, and presence of methoxy groups on the antiproliferative activity of [CuCl2(Ln)]. Their antiproliferative effect was analyzed in ovarian (A2780) and colorectal (HCT116) carcinomas and colorectal carcinoma resistant to doxorubicin (HCT116-DoxR) cell lines and in normal human fibroblasts. Among all complexes, the 1- and 2-naphtyl substituted terpy Cu(II) complexes (Cu1a and Cu1b) showed the strongest cytotoxicity, namely, in HCT116-DoxR 2Dcells and were also capable of inducing the loss of cell viability in 3D HCT116-DoxR spheroids. Their intracellular localization, capability to increase reactive oxygen species (ROS), and interaction with DNA (nonintercalative mode) trigger oxidative DNA cleavage leading to cell death by apoptosis and autophagy. Cu1a and Cu1b do not show in vivo toxicity in a chicken embryo and can interact with bovine serum albumin (BSA).
Collapse
Affiliation(s)
- Katarzyna Choroba
- Institute
of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland
| | - Bartosz Zowiślok
- Institute
of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland
| | - Sławomir Kula
- Institute
of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland
| | - Barbara Machura
- Institute
of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland
| | - Anna M. Maroń
- Institute
of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland
| | - Karol Erfurt
- Department
of Chemical Organic Technology and Petrochemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland
| | - Cristiana Marques
- Associate
Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2819-516 Caparica, Portugal
- Departamento
de Ciências da Vida, NOVA School of Science and Technology, UCIBIO, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Sandra Cordeiro
- Associate
Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2819-516 Caparica, Portugal
- Departamento
de Ciências da Vida, NOVA School of Science and Technology, UCIBIO, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Pedro V. Baptista
- Associate
Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2819-516 Caparica, Portugal
- Departamento
de Ciências da Vida, NOVA School of Science and Technology, UCIBIO, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Alexandra R. Fernandes
- Associate
Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School
of Science and Technology, NOVA University
Lisbon, 2819-516 Caparica, Portugal
- Departamento
de Ciências da Vida, NOVA School of Science and Technology, UCIBIO, Campus de Caparica, 2829-516 Caparica, Portugal
| |
Collapse
|
40
|
Kamala K, Ganapathy D, Sivaperumal P. Advancements in Cancer Therapy: Mycoviruses and Their Oncolytic Potential. Cell Biochem Biophys 2024:10.1007/s12013-024-01608-y. [PMID: 39535660 DOI: 10.1007/s12013-024-01608-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Recent advancements in cancer research focus on reducing treatment side effects while enhancing efficacy against medication resistance and tumor antigen detection. Genetic therapies utilizing microbes like bacteria, fungi, and viruses have garnered attention, with mycoviruses emerging as promising candidates. Particularly, the smallest fungal virus, Myco-phage, exhibits oncolytic properties by lysing cancer cells in the mouth, oral cavity, head, and neck without adverse effects. Genetically Modified Myco-phage (GmMP) adapts quickly to target cancer cells through cell membrane damage, inducing apoptosis and dendritic cell activation. Additionally, GmMP inhibits angiogenesis and modulates immune responses via CAR cells and immune checkpoints, potentially transforming cancer treatment paradigms with enhanced specificity and efficacy.
Collapse
Affiliation(s)
- Kannan Kamala
- Marine Microbial Research Lab, Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
- Centre for Marine and Aquatic Research (CMAR), Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 602105, India
| | - Dhanraj Ganapathy
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Pitchiah Sivaperumal
- Centre for Marine and Aquatic Research (CMAR), Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 602105, India.
- Marine Biomedical Research Lab & Environmental Toxicology Unit, Cellular and Molecular Research Centre, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India.
| |
Collapse
|
41
|
Tang N, Deng W, Wu Y, Deng Z, Wu X, Xiong J, Zhao Q. Single-Cell Spatial-Temporal Analysis of ZNF451 in Mediating Drug Resistance and CD8 + T Cell Dysfunction. RESEARCH (WASHINGTON, D.C.) 2024; 7:0530. [PMID: 39534688 PMCID: PMC11555180 DOI: 10.34133/research.0530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/02/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
Cisplatin is widely used to treat osteosarcoma, but recurrent cases often develop resistance, allowing the disease to progress and complicating clinical management. This study aimed to elucidate the immune microenvironment of osteosarcoma, providing insights into the mechanisms of recurrence and identifying potential therapeutic strategies. By analyzing multiple single-cell and bulk RNA-sequencing datasets, we discovered that the SUMOylation-related gene ZNF451 promotes osteosarcoma recurrence and alters its immune microenvironment. ZNF451 was found to importantly enhance the growth, migration, and invasion of resistant cells while also reducing their sensitivity to cisplatin and lowering their apoptosis rate. Moreover, our data indicated that ZNF451 plays a crucial role in bone resorption and epithelial-mesenchymal transition. ZNF451 also regulates CD8+ T cell function, leading to their exhaustion and transition to the CD8T.EXH state. Additionally, β-cryptoxanthin has been identified as a potential therapeutic agent that inhibits osteosarcoma progression by targeting ZNF451. In summary, these findings highlight the critical role of ZNF451 in promoting osteosarcoma progression and underscore its potential as a therapeutic target and biomarker for osteosarcoma.
Collapse
Affiliation(s)
- Ning Tang
- Department of Orthopaedics, Third Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Department of Orthopaedics, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Woding Deng
- Xiangya School of Medicine,
Central South University, Changsha, Hunan, China
| | - Yupeng Wu
- Department of Spine Surgery,
First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Zhixuan Deng
- Institute of Cell Biology, Hengyang Medical School,
University of South China, Hengyang, Hunan, China
| | - Xin Wu
- Department of Spine Surgery, Third Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Jianbin Xiong
- Department of Orthopaedics, Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Qiangqiang Zhao
- Department of Hematology,
Liuzhou People’s Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
- Department of Hematology,
The Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| |
Collapse
|
42
|
Li L, Li L, Wang Y, Wu B, Guan Y, Chen Y, Zhao J. Integration of Machine Learning and Experimental Validation to Identify Anoikis-Related Prognostic Signature for Predicting the Breast Cancer Tumor Microenvironment and Treatment Response. Genes (Basel) 2024; 15:1458. [PMID: 39596658 PMCID: PMC11594124 DOI: 10.3390/genes15111458] [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: 10/21/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Anoikis-related genes (ANRGs) are crucial in the invasion and metastasis of breast cancer (BC). The underlying role of ANRGs in the prognosis of breast cancer patients warrants further study. Methods: The anoikis-related prognostic signature (ANRS) was generated using a variety of machine learning methods, and the correlation between the ANRS and the tumor microenvironment (TME), drug sensitivity, and immunotherapy was investigated. Moreover, single-cell analysis and spatial transcriptome studies were conducted to investigate the expression of prognostic ANRGs across various cell types. Finally, the expression of ANRGs was verified by RT-PCR and Western blot analysis (WB), and the expression level of PLK1 in the blood was measured by the enzyme-linked immunosorbent assay (ELISA). Results: The ANRS, consisting of five ANRGs, was established. BC patients within the high-ANRS group exhibited poorer prognoses, characterized by elevated levels of immune suppression and stromal scores. The low-ANRS group had a better response to chemotherapy and immunotherapy. Single-cell analysis and spatial transcriptomics revealed variations in ANRGs across cells. The results of RT-PCR and WB were consistent with the differential expression analyses from databases. NU.1025 and imatinib were identified as potential inhibitors for SPIB and PLK1, respectively. Additionally, findings from ELISA demonstrated increased expression levels of PLK1 in the blood of BC patients. Conclusions: The ANRS can act as an independent prognostic indicator for BC patients, providing significant guidance for the implementation of chemotherapy and immunotherapy in these patients. Additionally, PLK1 has emerged as a potential blood-based diagnostic marker for breast cancer patients.
Collapse
Affiliation(s)
- Longpeng Li
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Longhui Li
- School of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing 100191, China
| | - Yaxin Wang
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Baoai Wu
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Yue Guan
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Yinghua Chen
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Jinfeng Zhao
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| |
Collapse
|
43
|
Zhao H, Jin Z, Li J, Fang J, Wu W, Fang JF. Novel insights of disulfidptosis-mediated immune microenvironment regulation in atherosclerosis based on bioinformatics analyses. Sci Rep 2024; 14:27336. [PMID: 39521794 PMCID: PMC11550432 DOI: 10.1038/s41598-024-78392-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Atherosclerosis (AS) is the leading cause of coronary heart disease, which is the primary cause of death worldwide. Recent studies have identified disulfidptosis as a new type of cell death that may be involved in onset and development of many diseases. However, the role of disulfidptosis in AS is not clear. In this study, bioinformatics analysis and experiments in vivo and in vitro were performed to evaluate the potential relationship between disulfidptosis and AS. AS-related sequencing data were obtained from the Gene Expression Omnibus (GEO). Bioinformatics techniques were used to evaluate differentially expressed genes (DEGs) associated with disulfidptosis-related AS. Hub genes were screened using least absolute shrinkage and selection operator (LASSO) and random forests (RF) methods. In addition, we established a foam cell model in vitro and an AS mouse model in vivo to verify the expressions of hub genes. In addition, we constructed a diagnostic nomogram with hub genes to predict progression of AS. Finally, the consensus clustering method was used to establish two different subtypes, and associations between subtypes and immunity were explored. As the results, 9 disulfidptosis-related AS DEGs were identified from GSE28829 and GSE43292 datasets. Evaluation of DEGs using LASSO and RF methods resulted in identification of 4 hub genes (CAPZB, DSTN, MYL6, PDLIM1), which were analyzed for diagnostic value using ROC curve analysis and verified in vitro and in vivo. Furthermore, a nomogram including hub genes was established that accurately predicted the occurrence of AS. The consensus clustering algorithm was used to separate patients with early atherosclerotic plaques and patients with advanced atherosclerotic plaques into two disulfidptosis subtypes. Cluster B displayed higher levels of infiltrating immune cells, which indicated that patients in cluster B may have a positive immune response for progression of AS. In summary, disulfidptosis-related genes including CAPZB, DSTN, MYL6, and PDLIM1 may be diagnostic markers and therapeutic targets for AS. In addition, these genes are closely related to immune cells, which may inform immunotherapy for AS.
Collapse
Affiliation(s)
- Huanyi Zhao
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Zheng Jin
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Junlong Li
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Junfeng Fang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Wei Wu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| | - J F Fang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| |
Collapse
|
44
|
Ge Y, Jiang L, Yang C, Dong Q, Tang C, Xu Y, Zhong X. Interactions between tumor-associated macrophages and regulated cell death: therapeutic implications in immuno-oncology. Front Oncol 2024; 14:1449696. [PMID: 39575419 PMCID: PMC11578871 DOI: 10.3389/fonc.2024.1449696] [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: 06/15/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
Tumor-associated macrophages (TAMs) play a pivotal role in sculpting the tumor microenvironment and influencing cancer progression, particularly through their interactions with various forms of regulated cell death (RCD), including apoptosis, pyroptosis, ferroptosis, and necroptosis. This review examines the interplay between TAMs and these RCD pathways, exploring the mechanisms through which they interact to promote tumor growth and advancement. We examine the underlying mechanisms of these intricate interactions, emphasizing their importance in cancer progression and treatment. Moreover, we present potential therapeutic strategies for targeting TAMs and manipulating RCD to enhance anti-tumor responses. These strategies encompass reprogramming TAMs, inhibiting their recruitment, and selectively eliminating them to enhance anti-tumor functions, alongside modulating RCD pathways to amplify immune responses. These insights offer a novel perspective on tumor biology and provide a foundation for the development of more efficacious cancer therapies.
Collapse
Affiliation(s)
- Yifei Ge
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lixue Jiang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chengru Yang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qingfu Dong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chengwu Tang
- Department of Hepatopancreatobiliary Surgery, Huzhou Key Laboratory of Translational Medicine, First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Hepatopancreatobiliary Surgery, Huzhou Key Laboratory of Translational Medicine, First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
45
|
He Y, Wang X. A comprehensive investigation of associations between cell death pathways and molecular and clinical features in pan-cancer. Clin Transl Oncol 2024:10.1007/s12094-024-03769-x. [PMID: 39487950 DOI: 10.1007/s12094-024-03769-x] [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: 09/13/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Regulated cell death (RCD) pathways play significant roles in tumorigenesis. However, systematic investigation into correlations between RCD and various molecular and clinical features, particularly anti-tumor immunity and immunotherapy response in pan-cancer remains lacking. METHODS Using the single-sample gene set enrichment analysis, we quantified the activities of six RCD pathways (apoptosis, autophagy, ferroptosis, cuproptosis, necroptosis, and pyroptosis) in each cancer specimen. Then, we explored associations of these six RCD pathways with tumor immunity, genomic instability, tumor phenotypes and clinical features, and responses to immunotherapy and targeted therapies in pan-cancer by statistical analyses. RESULTS Our results showed that the RCD (except autophagy) activities were oncogenic signatures, as evidenced by their hyperactivation in late stage or metastatic cancer patients, positive correlations with tumor proliferation, stemness, genomic instability and intratumor heterogeneity, and correlation with worse survival outcomes in cancer. In contrast, autophagy was a tumor suppressive signature as its associations with molecular and clinical features in cancer shows an opposite pattern compared to the other RCD pathways. Furthermore, heightened RCD (except cuproptosis) activities were correlated with increased sensitivity to immune checkpoint inhibitors. Additionally, elevated activities of pyroptosis, autophagy, cuproptosis and necroptosis were associated with increased drug sensitivity in a broad spectrum of anti-tumor targeted therapies, while the elevated activity of ferroptosis was correlated with decreased sensitivity to numerous targeted therapies. CONCLUSION RCD (except autophagy) activities correlate with unfavorable cancer prognosis, while the autophagy activity correlate with favorable clinical outcomes. RCD (except cuproptosis) activities are positive biomarkers for anti-tumor immunity and immunotherapy response.
Collapse
Affiliation(s)
- Yin He
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China.
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
46
|
Rajan SS, Chandran R, Abrahamse H. Advancing Photodynamic Therapy with Nano-Conjugated Hypocrellin: Mechanisms and Clinical Applications. Int J Nanomedicine 2024; 19:11023-11038. [PMID: 39502636 PMCID: PMC11537162 DOI: 10.2147/ijn.s486014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/03/2024] [Indexed: 11/08/2024] Open
Abstract
Hypocrellin-based photodynamic therapy (PDT) is developing as a viable cancer therapeutic option, especially when enhanced by nanoconjugation. This review investigates the methods by which nano-conjugated hypocrellin enhances therapeutic efficacy and precision when targeting cancer cells. These nanoconjugates encapsulate or covalently bind hypocrellin photosensitizers (PSs), allowing them to accumulate preferentially in malignancies. When activated by light, the nanoconjugates produce singlet oxygen and other reactive oxygen species (ROS), resulting in oxidative stress that selectively destroys cancer cells while protecting healthy tissues. We look at how they can be used to treat a variety of cancers. Clinical and preclinical studies show that they have advantages such as increased water solubility, improved tumor penetration, longer circulation times, and tailored delivery, all of which contribute to fewer off-target effects and overall toxicity. Ongoing research focuses on improving these nanoconjugates for better tumor targeting, drug release kinetics, and overcoming biological obstacles. Furthermore, the incorporation of developing technologies such as stimuli-responsive nanocarriers and combination therapies opens exciting opportunities for enhancing hypocrellin-based PDT. In conclusion, the combination of hypocrellin and nanotechnology constitutes a significant approach to cancer treatment, increasing the efficacy and safety of PDT. Future research will seek to create conjugates including hypocrellin, herceptin, and gold nanoparticles to induce apoptosis in human breast cancer cells in vitro, opening possibilities for therapeutic applications.
Collapse
Affiliation(s)
- Sheeja S Rajan
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Rahul Chandran
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
47
|
Zhang L, Xie A, Ma J, Liu H, Zeng C. Unveiling Cuproptosis: Mechanistic insights, roles, and leading advances in oncology. Biochim Biophys Acta Rev Cancer 2024; 1879:189180. [PMID: 39276875 DOI: 10.1016/j.bbcan.2024.189180] [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: 05/17/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024]
Abstract
Copper, a vital micronutrient, performs essential functions in numerous biological settings. Its disrupted metabolism is implicated in both the initiation of tumors and therapeutic interventions for cancer, underscoring the critical necessity of preserving copper homeostasis. Cuproptosis, a regulated cell death (RCD) modulated by copper, is activated in response to elevated copper concentrations, prompting an investigation into its implication in oncogenesis. Within this review, an exploration is conducted into copper dynamics and homeostasis maintenance within cells. Furthermore, it delves into the mechanisms underlying cuproptosis and its interplay with signaling pathways implicated in cancer. The potential synergy between cuproptosis and ferroptosis and its impact on tumor immunomodulation is discussed. Additionally, promising avenues for addressing cuproptosis in cancer involve assessing the utility of copper chelators and ionophores. By addressing pressing questions surrounding cuproptosis and outlining its pivotal role in cancer pathogenesis and treatment, this review propounds targeting cuproptosis as a promising frontier in antitumor therapy, potentially revolutionizing cancer treatment strategies.
Collapse
Affiliation(s)
- Limei Zhang
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Aihui Xie
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Jingxian Ma
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Huilin Liu
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen 518110, China.
| |
Collapse
|
48
|
Radha G, Pragyandipta P, Naik PK, Lopus M. The mode of action of sorafenib in MDA-MB-231 breast carcinoma cells involves components of apoptotic, necroptotic and autophagy-dependent cell death pathways. Exp Cell Res 2024; 443:114313. [PMID: 39486634 DOI: 10.1016/j.yexcr.2024.114313] [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: 08/16/2024] [Revised: 10/23/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
We report the identification of an interesting mode of action by sorafenib (SF) (Nexavar) in triple-negative breast adenocarcinoma MDA-MB-231 cells. The dying cells presented features of apoptosis, such as externalization of phosphatidylserine and cleaved caspase-3, and autophagy-mediated cell death, such as formation of autophagosomes and autolysosomes, the overexpression of LC3-II, and the presence of LAMP1-positive vacuoles, while displaying insufficient autophagic flux. Components of endoplasmic reticulum stress (ER stress; PERK and CHOP) and of necroptosis (p-MLKL) were also elevated considerably. Investigating potential target proteins that could modulate this form of cell death, we next investigated the role of tubulin disruption, which is known to induce necroptosis, apoptosis, and autophagy-dependent cell death. Interactions of SF with purified tubulin were investigated in detail using a combination of cellular and biophysical assays, transmission electron microscopy, and computer simulations. A marked reduction in the intrinsic tryptophan fluorescence of tubulin, a concentration-dependent elevation of anilinonaphthalene sulfonate-tubulin complex fluorescence, electron micrographs of deformed in vitro-assembled microtubules, and disrupted and hyper-stabilized cellular microtubules evinced the ability of SF to target tubulin and disrupt cellular microtubules. Molecular docking and molecular dynamic simulations positioned the drug between the α and β subunits of tubulin with considerable stability (ΔGbind, -31.43 kcal/mol), suggesting that drug-induced perturbation of tubulin could contribute to this mode of cell death.
Collapse
Affiliation(s)
- Gudapureddy Radha
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, Mumbai, 400098, India
| | - Pratyush Pragyandipta
- Center of Excellence in Natural Products and Therapeutics, Department of Biotechnology and Bioinformatics, Sambalpur University, Sambalpur, 768019, Odisha, India
| | - Pradeep Kumar Naik
- Center of Excellence in Natural Products and Therapeutics, Department of Biotechnology and Bioinformatics, Sambalpur University, Sambalpur, 768019, Odisha, India
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, Mumbai, 400098, India.
| |
Collapse
|
49
|
Mi T, Kong X, Chen M, Guo P, He D. Inducing disulfidptosis in tumors:potential pathways and significance. MedComm (Beijing) 2024; 5:e791. [PMID: 39415848 PMCID: PMC11480524 DOI: 10.1002/mco2.791] [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: 06/05/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Regulated cell death (RCD) is crucial for the elimination of abnormal cells. In recent years, strategies aimed at inducing RCD, particularly apoptosis, have become increasingly important in cancer therapy. However, the ability of tumor cells to evade apoptosis has led to treatment resistance and relapse, prompting extensive research into alternative death processes in cancer cells. A recent study identified a novel form of RCD known as disulfidptosis, which is linked to disulfide stress. Cancer cells import cystine from the extracellular environment via solute carrier family 7 member 11 (SLC7A11) and convert it to cysteine using nicotinamide adenine dinucleotide phosphate (NADPH). When NADPH is deficient or its utilization is impaired, cystine accumulates, leading to the formation of disulfide bonds in the actin cytoskeleton, triggering disulfidptosis. Disulfidptosis reveals a metabolic vulnerability in tumors, offering new insights into cancer therapy strategies. This review provides a detailed overview of the mechanisms underlying disulfidptosis, the current research progress, and limitations. It also highlights innovative strategies for inducing disulfidptosis and explores the potential of combining these approaches with traditional cancer therapies, particularly immunotherapy, to expedite clinical translation.
Collapse
Affiliation(s)
- Tao Mi
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and DisordersChongqingP.R. China
- Chongqing Key Laboratory of Structural Birth Defect and ReconstructionChongqingP.R. China
| | - Xiangpan Kong
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and DisordersChongqingP.R. China
- Chongqing Key Laboratory of Structural Birth Defect and ReconstructionChongqingP.R. China
| | - Meiling Chen
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and DisordersChongqingP.R. China
- Chongqing Key Laboratory of Structural Birth Defect and ReconstructionChongqingP.R. China
| | - Peng Guo
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and DisordersChongqingP.R. China
- Chongqing Key Laboratory of Structural Birth Defect and ReconstructionChongqingP.R. China
- Institute of Basic Medicine and Cancer (IBMC)Chinese Academy of SciencesHangzhouP.R. China
| | - Dawei He
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and DisordersChongqingP.R. China
- Chongqing Key Laboratory of Structural Birth Defect and ReconstructionChongqingP.R. China
| |
Collapse
|
50
|
Güçlü E, Ayan İÇ, Çetinkaya S, Dursun HG, Vural H. Piceatannol induces caspase-dependent apoptosis by modulating intracellular reactive oxygen species/mitochondrial membrane potential and enhances autophagy in neuroblastoma cells. J Appl Toxicol 2024; 44:1714-1724. [PMID: 39004823 DOI: 10.1002/jat.4671] [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: 05/14/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024]
Abstract
The aim of this study was to evaluate the anticancer effects of piceatannol, a natural stilbenoid, on human neuroblastoma cells. In order to accomplish this goal, we performed various cellular assays, including the XTT cell proliferation assay for cell viability, colony formation assay for colony formation capacity, FITC Annexin V and cell death detection kit for apoptosis, matrigel invasion assay for invasion capacity, intracellular reactive oxygen species (ROS) red dye for intracellular ROS levels, TMRM staining method for mitochondrial membrane potential (MMP), and the CYTO-ID autophagy detection kit for autophagy. Furthermore, we analyzed the expression levels of genes associated with apoptosis and autophagy using RT-qPCR. Based on our findings, piceatannol exhibited cytotoxic effects on neuroblastoma cells. Besides, treatment with piceatannol at both 50 and 100 μM concentrations for 72 h decreased colony formation, induced apoptosis and autophagy, inhibited cell invasion, decreased MMP, and increased ROS levels in SH-SY5Y cells. In addition, we observed significant upregulation in the expression levels of CASP8, BECLIN, ATG5, ATG7, and MAPILC3A genes between the two doses. These results suggest that piceatannol enhances autophagic activity and induces caspase-dependent apoptosis, indicating its potential as a therapeutic agent against neuroblastoma cells.
Collapse
Affiliation(s)
- Ebru Güçlü
- Department of Basic Science and Health, Hemp Research Institute, Yozgat Bozok University, Yozgat, Turkey
| | - İlknur Çınar Ayan
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Sümeyra Çetinkaya
- Biotechnology Research Center, Field Crops Central Research Institute, Ankara, Türkiye
| | - Hatice Gül Dursun
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Hasibe Vural
- Department of Medical Biology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|