1
|
Namikawa Y, Kato Y, Hokura A, Homma-Takeda S, Suzuki M. Extensive iron accumulation in the digestive gland of Turbo sazae and characterization of iron distribution and chemical structure. Food Chem 2025; 485:144552. [PMID: 40318333 DOI: 10.1016/j.foodchem.2025.144552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/09/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Metal accumulation in marine invertebrates has attracted significant attention owing to its toxicity to human health. Although T. sazae is a major edible fish resource in Japan, metal accumulation in the soft body of T. sazae has not been examined. In this study, the metal concentrations, chemical forms, and distributions in soft tissues of T. sazae were characterized. We observed the extensive iron accumulation i.e. over 10,000 μg/g dw in the digestive gland of T. sazae. The iron accumulator in the digestive gland was ferritin, an iron storage protein. Analysis of elemental distribution revealed that ferritin in the digestive gland contains phosphorus, and that μm-sized brown granular cells were responsible for iron storage, with over 70,000 μg/g ww of iron accumulated at the most concentrated point. T. sazae probably contributes to the ocean's iron cycle by grazing on iron-rich algae and rocks and storing iron using unique ferritin.
Collapse
Affiliation(s)
- Yuto Namikawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan
| | - Yugo Kato
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan
| | - Akiko Hokura
- Department of Applied Chemistry, School of Engineering, Tokyo Denki University, 5 Senju-Asahicho, Adachi, Tokyo 120-8551, Japan
| | - Shino Homma-Takeda
- Institute for Radiological Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Michio Suzuki
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan.
| |
Collapse
|
2
|
Kwon J, Kim SW, Hong S, Choi A, Choi S, Park MK, Kim HJ. MCOLN1/TRPML1-MCOLN3/TRPML3 heteromer and its coupling to Ca 2+ sensor SYT5 regulates autophagosome-lysosome fusion in a PtdIns4P-dependent manner. Autophagy 2025:1-17. [PMID: 40413756 DOI: 10.1080/15548627.2025.2510846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 05/16/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025] Open
Abstract
Macroautophagy/autophagy progresses through Ca2+-dependent multiple fusion events. Recently, we reported that the intracellular Ca2+ channel MCOLN3/TRPML3 provides Ca2+ for membrane fusion during autophagosome formation as a downstream effector of phosphatidylinositol-3-phosphate (PtdIns3P). However, the molecular mechanism of Ca2+ signaling in the late stage of autophagy remains unknown. Here, we show that the MCOLN1/TRPML1-MCOLN3/TRPML3 heteromer is the Ca2+ provider for autophagosome-lysosome fusion. MCOLN1-MCOLN3 functions downstream of PtdIns4P to release Ca2+ from autophagosomes, and the Ca2+ signal via PtdIns4P-MCOLN1-MCOLN3 is decoded by the Ca2+ sensor SYT5 (synaptotagmin 5). The binding of Ca2+ and PtdIns4P to SYT5 is critical for autophagosome-lysosome fusion by forming a fusion complex. Collectively, these results reveal that PtdIns4P-MCOLN1-MCOLN3-SYT5 constitutes the Ca2+ signaling complex in autophagosome-lysosome fusion and that MCOLN3 also regulates the late stage of autophagy through heteromerization with MCOLN1 in a phosphoinositide (PI)-dependent manner.Abbreviations: ATG, autophagy related; CPA, cyclopiazonic acid; DN, dominant-negative; GPN, glycyl-L-phenylalanine-beta-naphthylamide; KO, knockout; NH4Cl, ammonium chloride; PtdIns3K, phosphatidylinositol 3-kinase; PtdIns3P, phosphatidylinositol-3-phosphate; PI, phosphoinositide; SYT5, synaptotagmin 5; tfLC3, mRFP-GFP tandem fluorescent-tagged LC3; WT, wild-type.
Collapse
Affiliation(s)
- Jin Kwon
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - So Woon Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Seokwoo Hong
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Areum Choi
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Suzi Choi
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Myoung Kyu Park
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun Jin Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| |
Collapse
|
3
|
Gomes GMDA, Xu M, Syeda AKR, Raudonis R, Almasi S, Vijayan VV, Gujar S, Dong X, Cheng Z, Pulinilkunnil T, El Hiani Y. Targeting TRPML3 inhibits proliferation and invasion, and enhances doxorubicin sensitivity by disrupting lysosomal acidification and mitochondrial function in triple-negative breast cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119979. [PMID: 40348344 DOI: 10.1016/j.bbamcr.2025.119979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 04/22/2025] [Accepted: 04/27/2025] [Indexed: 05/14/2025]
Abstract
TNBC remains the most aggressive and therapy-resistant type of breast cancer, for which efficient targeted therapies have not been developed yet. Here, we identified TRPML3 (ML3) as a potential therapeutic target in TNBC. Our data showed that ML3 is significantly upregulated in TNBC cells compared with nontumorigenic control cells. ML3 knockdown (KD) impairs TNBC cell proliferation by inducing cell cycle arrest and caspase-dependent apoptosis. ML3 KD also inhibits TNBC cell migration and invasion. Mechanistically, ML3 KD reduces lysosomal number and enhances lysosomal acidification, which in turn activates mTORC1, thereby inhibiting autophagy initiation and flux. This disruption negatively impacts mitochondrial function, as evidenced by reduced ATP production, increased ROS and NO production, and mitochondrial fragmentation. Importantly, ML3 KD enhances TNBC cell sensitivity to doxorubicin and paclitaxel. The finding suggests that targeting ML3 disrupts lysosomal and mitochondrial homeostasis and enhance chemosensitivity, presenting ML3 as a potential therapeutic vulnerability in TNBC enhancing chemosensitivity.
Collapse
Affiliation(s)
| | - Mengnan Xu
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada.
| | | | - Renee Raudonis
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | | | - Vishnu Vijay Vijayan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Shashi Gujar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Xianping Dong
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Zhenyu Cheng
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | | | - Yassine El Hiani
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
4
|
Kondratskyi A, Bazzone A, Rapedius M, Zerlotti R, Masson B, Sadanandan NP, Parker JL, Santinho A, Moutia M, Thiam AR, Kemp A, Seibertz F, Murciano N, Friis S, Becker N, Obergrussberger A, Barthmes M, George C, George M, Dalrymple D, Gasnier B, Newstead S, Grimm C, Fertig N. Lysosomal Ion Channels and Transporters: Recent Findings, Therapeutic Potential, and Technical Approaches. Bioelectricity 2025; 7:29-57. [PMID: 40342936 PMCID: PMC12056583 DOI: 10.1089/bioe.2025.0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025] Open
Abstract
In recent years, there has been a growing interest in lysosomal ion channels and transporters due to their critical role in maintaining lysosomal function and their involvement in a variety of diseases, particularly lysosomal storage diseases, cancer, and neurodegenerative disorders. Recent advancements in research techniques, including manual and automated patch clamp (APC) electrophysiology, solid-supported membrane-based electrophysiology (SSME), and fluorescence-based ion imaging, have further enhanced our ability to investigate lysosomal ion channels and transporters in both physiological and pathological conditions, spurring drug discovery efforts. Several pharmaceutical companies are now developing therapies aimed at modulating these channels and transporters to improve lysosomal function in disease. Small molecules targeting channels like transient receptor potential mucolipin (TRPML) 1 and TMEM175, as well as drugs modulating lysosomal pH, are currently in preclinical and clinical development. This review provides an overview of the role of lysosomal ion channels and transporters in health and disease, highlights the cutting-edge techniques used to study them, and discusses the therapeutic potential of targeting these channels and transporters in the treatment of various diseases. Furthermore, in addition to summarizing recent discoveries, we contribute novel functional data on cystinosin, TRPML1, and two-pore channel 2 (TPC2), utilizing both SSME and APC approaches.
Collapse
Affiliation(s)
| | | | | | | | | | - Nidish Ponath Sadanandan
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
| | - Joanne L. Parker
- Department of Biochemistry, University of Oxford, Oxford, UK
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | | | | | - Abdou Rachid Thiam
- Laboratoire de Physique de l’École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Arlene Kemp
- SB Drug Discovery a Sygnature Discovery Business, West of Scotland Science Park, Glasgow, UK
| | | | | | | | | | | | | | | | | | - David Dalrymple
- SB Drug Discovery a Sygnature Discovery Business, West of Scotland Science Park, Glasgow, UK
| | - Bruno Gasnier
- Saints-Pères Paris Institute for the Neurosciences, Université Paris Cité, Centre National de la Recherche Scientifique, Paris, France
| | - Simon Newstead
- Department of Biochemistry, University of Oxford, Oxford, UK
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
- Immunology, Infection and Pandemic Research IIP, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Munich/Frankfurt, Germany
| | | |
Collapse
|
5
|
Du Y, Kowada T, Sung E, Liu R, Soloviev A, Matsui T, Mizukami S. Quantification of lysosomal labile Zn 2+ and monitoring of Zn 2+ efflux using a small-molecule-protein hybrid fluorescent probe. J Inorg Biochem 2025; 264:112811. [PMID: 39674094 DOI: 10.1016/j.jinorgbio.2024.112811] [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: 08/25/2024] [Revised: 11/22/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Lysosomal labile Zn2+ levels have been unclear. By targeting a small-molecule fluorescent Zn2+ probe, ZnDA-3H, to lysosomes via VAMP7-Halo, the lysosomal labile Zn2+ concentration was determined to be 1.9 nM in HeLa cells. Furthermore, ZnDA-3H enabled direct visualization of the Zn2+ efflux from the lysosomes to cytosol upon TRPMLs activation.
Collapse
Affiliation(s)
- Yuyin Du
- Department of Chemistry, Faculty of Science, Tohoku University, 6-3 Aramaki Aza-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Toshiyuki Kowada
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan; Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan; Department of Chemistry, Graduate School of Science, Tohoku University, 6-3 Aramaki Aza-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| | - EunHye Sung
- Department of Chemistry, Graduate School of Science, Tohoku University, 6-3 Aramaki Aza-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Rong Liu
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Andrei Soloviev
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Toshitaka Matsui
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan; Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan; Department of Chemistry, Graduate School of Science, Tohoku University, 6-3 Aramaki Aza-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Shin Mizukami
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan; Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan; Department of Chemistry, Graduate School of Science, Tohoku University, 6-3 Aramaki Aza-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| |
Collapse
|
6
|
Bai J, Wan Z, Zhou W, Wang L, Lou W, Zhang Y, Jin H. Global trends and emerging insights in BRAF and MEK inhibitor resistance in melanoma: a bibliometric analysis. Front Mol Biosci 2025; 12:1538743. [PMID: 39897423 PMCID: PMC11782018 DOI: 10.3389/fmolb.2025.1538743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Objective This study aims to perform a comprehensive bibliometric analysis of global research on BRAF and MEK inhibitor resistance in melanoma, identifying key research trends, influential contributors, and emerging themes from 2003 to 2024. Methods A systematic search was conducted in the Web of Science Core Collection (WoSCC) database to retrieve publications related to BRAF and MEK inhibitor resistance from 1 January 2003, to 1 September 2024. Bibliometric analyses, including publication trends, citation networks, and keyword co-occurrence patterns, were performed using VOSviewer and CiteSpace. Collaborative networks, co-cited references, and keyword burst analyses were mapped to uncover shifts in research focus and global cooperation. Results A total of 3,503 documents, including 2,781 research articles and 722 review papers, were analyzed, highlighting significant growth in this field. The United States, China, and Italy led in publication volume and citation impact, with Harvard University and the University of California System among the top contributing institutions. Research output showed three phases of growth, peaking in 2020. Keyword and co-citation analyses revealed a transition from early focus on BRAF mutations and MAPK pathway activation to recent emphasis on immunotherapy, combination therapies, and non-apoptotic cell death mechanisms like ferroptosis and pyroptosis. These trends reflect the evolving priorities and innovative approaches shaping the field of resistance to BRAF and MEK inhibitors in melanoma. Conclusion Research on BRAF and MEK inhibitor resistance has evolved significantly. This analysis provides a strategic framework for future investigations, guiding the development of innovative, multi-modal approaches to improve treatment outcomes for melanoma patients.
Collapse
Affiliation(s)
- Jianhao Bai
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongqi Wan
- Department of Ophthalmology, Shanghai Tenth People’s Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai, China
| | - Wanru Zhou
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lijun Wang
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Lou
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yao Zhang
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haiying Jin
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Liu Z, Gan Y, Shen Z, Cai S, Wang X, Li Y, Li X, Fu H, Chen J, Li N. Role of copper homeostasis and cuproptosis in heart failure pathogenesis: implications for therapeutic strategies. Front Pharmacol 2025; 15:1527901. [PMID: 39850564 PMCID: PMC11754225 DOI: 10.3389/fphar.2024.1527901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Copper is an essential micronutrient involved in various physiological processes in various cell types. Consequently, dysregulation of copper homeostasis-either excessive or deficient-can lead to pathological changes, such as heart failure (HF). Recently, a new type of copper-dependent cell death known as cuproptosis has drawn increasing attention to the impact of copper dyshomeostasis on HF. Notably, copper dyshomeostasis was associated with the occurrence of HF. Hence, this review aimed to investigate the biological processes involved in copper uptake, transport, excretion, and storage at both the cellular and systemic levels in terms of cuproptosis and HF, along with the underlying mechanisms of action. Additionally, the role of cuproptosis and its related mitochondrial dysfunction in HF pathogenesis was analyzed. Finally, we reviewed the therapeutic potential of current drugs that target copper metabolism for treating HF. Overall, the conclusions of this review revealed the therapeutic potential of copper-based therapies that target cuproptosis for the development of strategies for the treatment of HF.
Collapse
Affiliation(s)
- Zhichao Liu
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Yongkang Gan
- Department of Vascular Surgery, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Zhen Shen
- Department of Clinical Laboratory, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Siqi Cai
- College of Art, Nanjing University of Information Science and Technology, Nanjing, Jiangsu, China
| | - Xizhen Wang
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Yong Li
- Experimental Center for Medical Research, Shandong Second Medical University, Weifang, Shandong, China
| | - Xiaofeng Li
- Department of Cardiovascular, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huanjie Fu
- Department of Cardiovascular, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinhong Chen
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Ningcen Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
8
|
Feng X, Cai W, Li Q, Zhao L, Meng Y, Xu H. Activation of lysosomal Ca2+ channels mitigates mitochondrial damage and oxidative stress. J Cell Biol 2025; 224:e202403104. [PMID: 39500490 PMCID: PMC11540856 DOI: 10.1083/jcb.202403104] [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: 03/17/2024] [Revised: 09/06/2024] [Accepted: 10/15/2024] [Indexed: 11/09/2024] Open
Abstract
Elevated levels of plasma-free fatty acids and oxidative stress have been identified as putative primary pathogenic factors in endothelial dysfunction etiology, though their roles are unclear. In human endothelial cells, we found that saturated fatty acids (SFAs)-including the plasma-predominant palmitic acid (PA)-cause mitochondrial fragmentation and elevation of intracellular reactive oxygen species (ROS) levels. TRPML1 is a lysosomal ROS-sensitive Ca2+ channel that regulates lysosomal trafficking and biogenesis. Small-molecule agonists of TRPML1 prevented PA-induced mitochondrial damage and ROS elevation through activation of transcriptional factor EB (TFEB), which boosts lysosome biogenesis and mitophagy. Whereas genetically silencing TRPML1 abolished the protective effects of TRPML1 agonism, TRPML1 overexpression conferred a full resistance to PA-induced oxidative damage. Pharmacologically activating the TRPML1-TFEB pathway was sufficient to restore mitochondrial and redox homeostasis in SFA-damaged endothelial cells. The present results suggest that lysosome activation represents a viable strategy for alleviating oxidative damage, a common pathogenic mechanism of metabolic and age-related diseases.
Collapse
Affiliation(s)
- Xinghua Feng
- New Cornerstone Science Laboratory and Liangzhu Laboratory, The Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Weijie Cai
- New Cornerstone Science Laboratory and Liangzhu Laboratory, The Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Qian Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Liding Zhao
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaping Meng
- New Cornerstone Science Laboratory and Liangzhu Laboratory, The Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Haoxing Xu
- New Cornerstone Science Laboratory and Liangzhu Laboratory, The Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
9
|
Li Y, Zhang H, Yang F, Zhu D, Chen S, Wang Z, Wei Z, Yang Z, Jia J, Zhang Y, Wang D, Ma M, Kang X. Mechanisms and therapeutic potential of disulphidptosis in cancer. Cell Prolif 2025; 58:e13752. [PMID: 39354653 DOI: 10.1111/cpr.13752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
SLC7A11 plays a pivotal role in tumour development by facilitating cystine import to enhance glutathione synthesis and counteract oxidative stress. Disulphidptosis, an emerging form of cell death observed in cells with high expression of SLC7A11 under glucose deprivation, is regulated through reduction-oxidation reactions and disulphide bond formation. This process leads to contraction and collapse of the F-actin cytoskeleton from the plasma membrane, ultimately resulting in cellular demise. Compared to other forms of cell death, disulphidptosis exhibits distinctive characteristics and regulatory mechanisms. This mechanism provides novel insights and innovative strategies for cancer treatment while also inspiring potential therapeutic approaches for other diseases. Our review focuses on elucidating the molecular mechanism underlying disulphidptosis and its connection with the actin cytoskeleton, identifying alternative metabolic forms of cell death, as well as offering insights into disulphidptosis-based cancer therapy. A comprehensive understanding of disulphidptosis will contribute to our knowledge about fundamental cellular homeostasis and facilitate the development of groundbreaking therapies for disease treatment.
Collapse
Affiliation(s)
- Yanhu Li
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Haijun Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
- The Second People's Hospital of Gansu Province, Lanzhou, PR China
| | - Fengguang Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Daxue Zhu
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Shijie Chen
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhaoheng Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Ziyan Wei
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhili Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Jingwen Jia
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Yizhi Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Dongxin Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Mingdong Ma
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Xuewen Kang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| |
Collapse
|
10
|
Shi Q, Yang Z, Yang H, Xu L, Xia J, Gu J, Chen M, Wang Y, Zhao X, Liao Z, Mou Y, Gu X, Xie T, Sui X. Targeting ion channels: innovative approaches to combat cancer drug resistance. Theranostics 2025; 15:521-545. [PMID: 39744692 PMCID: PMC11671388 DOI: 10.7150/thno.103384] [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: 09/07/2024] [Accepted: 10/21/2024] [Indexed: 01/11/2025] Open
Abstract
Ion channels, as functional molecules that regulate the flow of ions across cell membranes, have emerged as a promising target in cancer therapy due to their pivotal roles in cell proliferation, metastasis, apoptosis, drug resistance, and so on. Recently, increasing evidence suggests that dysregulation of ion channels is a common characteristic of cancer cells, contributing to their survival and the resistance to conventional therapies. For example, the aberrant expression of sodium (Na+) and potassium ion (K+) channels is significantly correlated with the sensitivity of chemotherapy drugs. The endogenous calcium (Ca2+) channels contribute to the acquired resistance of osimertinib in epidermal growth factor receptor (EGFR) mutant non-small cell lung cancer cell lines. Ferrous ions (Fe2+) enhance the sensitivity of breast cancer cells to doxorubicin treatment. Preclinical models have also demonstrated the effect of specific ion channel blockers or modulators on anticancer drug resistance. This review describes the current understanding about the interaction between ion channels and the therapeutic efficacy of anticancer drugs. Then, the therapeutic potential of ion channel blockers or modulators in enhancing the sensitivity or overcoming the resistance of cancer cells to anticancer therapies is discussed. Targeting ion channels will hopefully offer a novel and promising strategy for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Qian Shi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zijing Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Huan Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Lihui Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jing Xia
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jie Gu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengting Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yan Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaohong Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zehua Liao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yiping Mou
- General Surgery, Cancer Center, Department of Gastrointestinal-Pancreatic Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical University, Hangzhou, Zhejiang, China
| | - Xidong Gu
- Department of Breast Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Li T, Yu C. Metal-Dependent Cell Death in Renal Fibrosis: Now and in the Future. Int J Mol Sci 2024; 25:13279. [PMID: 39769044 PMCID: PMC11678559 DOI: 10.3390/ijms252413279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Renal fibrosis is a common final pathway underlying nearly almost all progressive kidney diseases. Metal ions are essential trace elements in organisms and are involved in important physiological activities. However, aberrations in intracellular metal ion metabolism may disrupt homeostasis, causing cell death and increasing susceptibility to various diseases. Accumulating evidence suggests a complex association between metal-dependent cell death and renal fibrosis. In this article, we provide a comprehensive overview of the specific molecular mechanisms of metal-dependent cell death and their crosstalk, up-to-date evidence supporting their role in renal fibrosis, therapeutic targeting strategies, and research needs, aiming to offer a rationale for future clinical treatment of renal fibrosis.
Collapse
Affiliation(s)
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|
12
|
Chen H, Tang T, Xue C, Liu X, Xi Z, Xie L, Kang R. Exploration and breakthrough in the mode of intervertebral disc cell death may lead to significant advances in treatments for intervertebral disc degeneration. J Orthop Surg Res 2024; 19:825. [PMID: 39639370 PMCID: PMC11619685 DOI: 10.1186/s13018-024-05280-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Low back pain caused by intervertebral disc degeneration (IDD) has emerged as a significant global public health concern, with far-reaching consequences for patients' quality of life and healthcare systems. Although previous research have revealed that the mechanisms of intervertebral disc cell apoptosis, pyroptosis and necroptosis can aggravate IDD damage by mediating inflammation and promoting extracellular matrix degradation, but they cannot explain the connection between different cell death mechanisms and ion metabolism disorders. The latest study shows that cell death mechanisms such as cellular senescence, ferroptosis, and cuproptosis, and PANopotosis have similar roles in the progression of intervertebral disc degeneration, but not exactly the same damage mechanism. This paper summarizes the effects of various cell death patterns on the disease progression of IDD, related molecular mechanisms and signaling pathways, providing new perspectives and potential clinical intervention strategies for the prevention and treatment of IDD.
Collapse
Affiliation(s)
- Heng Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Tian Tang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Congyang Xue
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Xin Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Zhipeng Xi
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Lin Xie
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
- Jiangsu Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| | - Ran Kang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
- Jiangsu Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
13
|
Abrahamian C, Ouologuem L, Tang R, Fröhlich T, Bartel K, Grimm C. TPC2: From Blond Hair to Melanoma? Cancers (Basel) 2024; 16:4065. [PMID: 39682251 DOI: 10.3390/cancers16234065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Two-pore channel 2 (TPC2) is expressed in endolysosomes throughout the human body, as well as in melanosomes of melanocytes. Melanocytes produce pigment, i.e., melanin, which determines hair and skin color but also protects from UV light. Extensive exposure to UV light is one of the major risk factors for the development of melanoma, which develops from pigment-producing cells, i.e., melanocytes. In recent years, several human TPC2 single nucleotide polymorphisms have been identified to increase the likelihood of carriers presenting with blond hair and hypopigmentation. These variants were all characterized as gain-of-function versions of TPC2. Vice versa, the loss of function of TPC2 increases melanin production and reduces cancer hallmarks such as proliferation, migration, invasion, tumor growth, and metastasis formation. The activity of TPC2 is controlled in a complex manner, with several endogenous ligands as well as a number of interacting proteins being involved. We will discuss here the role of TPC2 in pigmentation and its potential to impact melanoma development and progression and highlight recent findings on Rab7a as an enhancer of TPC2 activity.
Collapse
Affiliation(s)
- Carla Abrahamian
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig Maximilians University, 80336 Munich, Germany
| | - Lina Ouologuem
- Department of Pharmacy, Ludwig Maximilians University, 80539 Munich, Germany
| | - Rachel Tang
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig Maximilians University, 80336 Munich, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, Ludwig Maximilians University, 80539 Munich, Germany
| | - Karin Bartel
- Department of Pharmacy, Ludwig Maximilians University, 80539 Munich, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig Maximilians University, 80336 Munich, Germany
- Immunology, Infection and Pandemic Research IIP, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 80333 Munich, Germany
| |
Collapse
|
14
|
Zhang L, Deng R, Liu L, Du H, Tang D. Novel insights into cuproptosis inducers and inhibitors. Front Mol Biosci 2024; 11:1477971. [PMID: 39659361 PMCID: PMC11628392 DOI: 10.3389/fmolb.2024.1477971] [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: 09/27/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Cuproptosis is a new pattern of Cu-dependent cell death distinct from classic cell death pathways and characterized by aberrant lipoylated protein aggregation in TCA cycle, Fe-S cluster protein loss, HSP70 elevation, proteotoxic and oxidative stress aggravation. Previous studies on Cu homeostasis and Cu-induced cell death provide a great basis for the discovery of cuproptosis. It has gradually gathered enormous research interests and large progress has been achieved in revealing the metabolic pathways and key targets of cuproptosis, due to its role in mediating some genetic, neurodegenerative, cardiovascular and tumoral diseases. In terms of the key targets in cuproptosis metabolic pathways, they can be categorized into three types: oxidative stress, mitochondrial respiration, ubiquitin-proteasome system. And strategies for developing cuproptosis inducers and inhibitors involved in these targets have been continuously improved. Briefly, based on the essential cuproptosis targets and metabolic pathways, this paper classifies some relevant inducers and inhibitors including small molecule compounds, transcription factors and ncRNAs with the overview of principle, scientific and medical application, in order to provide reference for the cuproptosis study and target therapy in the future.
Collapse
Affiliation(s)
- Ligang Zhang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Ruiting Deng
- Beijing Mercer United International Education Consulting Co., Ltd., Guangzhou, China
| | - Lian Liu
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Dongsheng Tang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| |
Collapse
|
15
|
He Y, Fan Y, Ahmadpoor X, Wang Y, Li ZA, Zhu W, Lin H. Targeting lysosomal quality control as a therapeutic strategy against aging and diseases. Med Res Rev 2024; 44:2472-2509. [PMID: 38711187 DOI: 10.1002/med.22047] [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: 08/19/2023] [Revised: 04/04/2024] [Accepted: 04/21/2024] [Indexed: 05/08/2024]
Abstract
Previously, lysosomes were primarily referred to as the digestive organelles and recycling centers within cells. Recent discoveries have expanded the lysosomal functional scope and revealed their critical roles in nutrient sensing, epigenetic regulation, plasma membrane repair, lipid transport, ion homeostasis, and cellular stress response. Lysosomal dysfunction is also found to be associated with aging and several diseases. Therefore, function of macroautophagy, a lysosome-dependent intracellular degradation system, has been identified as one of the updated twelve hallmarks of aging. In this review, we begin by introducing the concept of lysosomal quality control (LQC), which is a cellular machinery that maintains the number, morphology, and function of lysosomes through different processes such as lysosomal biogenesis, reformation, fission, fusion, turnover, lysophagy, exocytosis, and membrane permeabilization and repair. Next, we summarize the results from studies reporting the association between LQC dysregulation and aging/various disorders. Subsequently, we explore the emerging therapeutic strategies that target distinct aspects of LQC for treating diseases and combatting aging. Lastly, we underscore the existing knowledge gap and propose potential avenues for future research.
Collapse
Affiliation(s)
- Yuchen He
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yishu Fan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xenab Ahmadpoor
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yumin Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, China
| | - Weihong Zhu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
Liu J, Feng G. The causal relationship between trace element status and upper gastrointestinal ulcers: a Mendelian randomization study. Front Nutr 2024; 11:1443090. [PMID: 39539362 PMCID: PMC11557352 DOI: 10.3389/fnut.2024.1443090] [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: 06/03/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background This study aimed to investigate the bidirectional causal relationships between trace elements (such as zinc, magnesium, phosphate, and folate) and upper gastrointestinal ulcers (including gastric and duodenal ulcers). We utilized a two-sample Mendelian randomization (MR) analysis to achieve this. Methods We conducted a two-sample MR analysis using summary-level data from genome-wide association studies (GWAS) obtained from public genomics repositories. We utilized a range of MR methods, including inverse-variance weighted (IVW), MR-Egger, and weighted median methods, and conducted a meta-analysis to synthesize results across different datasets. To ensure the robustness of our findings, we performed extensive sensitivity analyses, including pleiotropy assessment, heterogeneity tests, and leave-one-out analysis. Results Our findings are significant, indicating a positive causal relationship between increased zinc levels and the risk of gastric ulcers. Moreover, magnesium and folate appear to offer potential protective effects against gastroduodenal ulcers (p < 0.05). The meta-analysis further supports the causal relationship between zinc and gastric ulcers (p < 0.05), confirming zinc's significant causal impact on this condition. Conclusion The study confirms a positive causal relationship between zinc and gastric ulcers and highlights the complexity of how trace elements regulate the progression of upper gastrointestinal ulcers. These results provide a scientific basis for dietary recommendations regarding trace element intake in clinical and public health practices. They also offer new insights into effective prevention and treatment strategies for gastric and duodenal ulcers.
Collapse
Affiliation(s)
- Jianwei Liu
- Department of Gastroenterology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Gege Feng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
17
|
Zhang L, Deng R, Guo R, Jiang Y, Guan Y, Chen C, Zhao W, Huang G, Liu L, Du H, Tang D. Recent progress of methods for cuproptosis detection. Front Mol Biosci 2024; 11:1460987. [PMID: 39297074 PMCID: PMC11408227 DOI: 10.3389/fmolb.2024.1460987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/21/2024] [Indexed: 09/21/2024] Open
Abstract
Varying from other identified cell death pathways, cuproptosis is a new type of regulated cell death characterized by excess Cu ions, abnormal aggregation of lipoylated proteins in TCA cycle, loss of Fe-S cluster proteins, upregulation of HSP70, leading to proteotoxic and oxidative stress. Cuproptosis is highly concerned by scientific community and as the field of cuproptosis further develops, remarkable progress has been made in the verification and mechanism of cuproptosis, and methods used to detect cuproptosis have been continuously improved. According to the characteristic changes of cuproptosis, techniques based on cell death verification, Cu content, morphology, molecular biology of protein levels of cuproptosis-related molecules and biochemical pathways of cuproptosis-related enzyme activity and metabolites of oxidative stress, lipoic acid, TCA cycle, Fe-S cluster proteins, oxidative phosphorylation, cell respiration intensity have been subject to cuproptosis verification and research. In order to further deepen the understanding of detecting cuproptosis, the principle and application of common cuproptosis detection methods are reviewed and categorized in cellular phenomena and molecular mechanism in terms of cell death, Cu content, morphology, molecular biology, biochemical pathways with a flow chart. All the indicating results have been displayed in response to the markers of cuproptosis, their advantages and limitations are summaried, and comparison of cuproptosis and ferroptosis detection is performed in this study. Our collection of methods for cuproptosis detection will provide a great basis for cuproptosis verification and research in the future.
Collapse
Affiliation(s)
- Ligang Zhang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Ruiting Deng
- Beijing Mercer United International Education Consulting Co., Ltd., Guangzhou, China
| | - Raoqing Guo
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yawen Jiang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Yichen Guan
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Caiyue Chen
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Wudi Zhao
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Guobin Huang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Lian Liu
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Dongsheng Tang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| |
Collapse
|
18
|
Mao C, Wang M, Zhuang L, Gan B. Metabolic cell death in cancer: ferroptosis, cuproptosis, disulfidptosis, and beyond. Protein Cell 2024; 15:642-660. [PMID: 38428031 PMCID: PMC11365558 DOI: 10.1093/procel/pwae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024] Open
Abstract
Cell death resistance represents a hallmark of cancer. Recent studies have identified metabolic cell death as unique forms of regulated cell death resulting from an imbalance in the cellular metabolism. This review discusses the mechanisms of metabolic cell death-ferroptosis, cuproptosis, disulfidptosis, lysozincrosis, and alkaliptosis-and explores their potential in cancer therapy. Our review underscores the complexity of the metabolic cell death pathways and offers insights into innovative therapeutic avenues for cancer treatment.
Collapse
Affiliation(s)
- Chao Mao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Min Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Li Zhuang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
19
|
Xiao Y, Yin J, Liu P, Zhang X, Lin Y, Guo J. Triptolide-induced cuproptosis is a novel antitumor strategy for the treatment of cervical cancer. Cell Mol Biol Lett 2024; 29:113. [PMID: 39198750 PMCID: PMC11360305 DOI: 10.1186/s11658-024-00623-4] [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: 03/19/2024] [Accepted: 07/17/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Cuproptosis is a unique copper-dependent form of cell death that is highly correlated with the metabolic state of cells. Triptolide exerts pharmacological activity by altering the regulation of metal ions. Cuproptosis is poorly understood in cancer, so in this study, we explored whether triptolide could induce cuproptosis in cervical cancer cells. METHODS The human cervical cancer cell lines HeLa and SiHa, which primarily rely on oxidative phosphorylation, were treated with triptolide. Cell viability, proliferation and migration, copper levels and cuproptosis-related protein levels were evaluated in these cell lines. The copper ion chelator tetrathiomolybdate (TTM) was administered to determine whether it could reverse the cuproptosis induced by triptolide. In addition, a nude mouse cervical cancer xenograft model was established to determine the effects of triptolide on cuproptosis in isolated tumor tissues. RESULTS The copper concentration increased with triptolide treatment. The levels of cuproptosis -related proteins, such as FDX1, LIAS, and DLAT, in the HeLa and SiHa cell lines decreased with triptolide treatment. XIAP, the target of triptolide, played a role in cuproptosis by regulating COMMD1. The level of copper exporters (ATP7A/B) decreased, but the level of the copper importer (CTR1) did not change with triptolide treatment. Furthermore, triptolide inhibited cervical cancer growth and induced cuproptosis in vivo. CONCLUSIONS In summary, we report a new antitumor mechanism by which triptolide disrupted intracellular copper homeostasis and induced cuproptosis in cervical cancer by regulating the XIAP/COMMD1/ATP7A/B axis.
Collapse
Affiliation(s)
- Yanxia Xiao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, NO.1 Da HuaRoad, DongDan, Beijing, 100730, People's Republic of China
| | - Jiameng Yin
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, NO.1 Da HuaRoad, DongDan, Beijing, 100730, People's Republic of China
| | - Pu Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, NO.1 Da HuaRoad, DongDan, Beijing, 100730, People's Republic of China
| | - Xin Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, NO.1 Da HuaRoad, DongDan, Beijing, 100730, People's Republic of China
| | - Yajun Lin
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, NO.1 Da HuaRoad, DongDan, Beijing, 100730, People's Republic of China.
| | - Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, NO.1 Da HuaRoad, DongDan, Beijing, 100730, People's Republic of China.
| |
Collapse
|
20
|
Trojani MC, Santucci-Darmanin S, Breuil V, Carle GF, Pierrefite-Carle V. Lysosomal exocytosis: From cell protection to protumoral functions. Cancer Lett 2024; 597:217024. [PMID: 38871244 DOI: 10.1016/j.canlet.2024.217024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024]
Abstract
Lysosomes are single membrane bounded group of acidic organelles that can be involved in a process called lysosomal exocytosis which leads to the extracellular release of their content. Lysosomal exocytosis is required for plasma membrane repair or remodeling events such as bone resorption, antigen presentation or mitosis, and for protection against toxic agents such as heavy metals. Recently, it has been showed that to fulfill this protective role, lysosomal exocytosis needs some autophagic proteins, in an autophagy-independent manner. In addition to these crucial physiological roles, lysosomal exocytosis plays a major protumoral role in various cancers. This effect is exerted through tumor microenvironment modifications, including extracellular matrix remodeling, acidosis, oncogenic and profibrogenic signals. This review provides a comprehensive overview of the different elements released in the microenvironment during lysosomal exocytosis, i.e. proteases, exosomes, and protons, and their effects in the context of tumor development and treatment.
Collapse
Affiliation(s)
- Marie-Charlotte Trojani
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; Service de Rhumatologie, CHU de Nice, Nice, France
| | - Sabine Santucci-Darmanin
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; CNRS, Paris, France
| | - Véronique Breuil
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; Service de Rhumatologie, CHU de Nice, Nice, France
| | - Georges F Carle
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; CNRS, Paris, France
| | - Valérie Pierrefite-Carle
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; INSERM, Paris, France.
| |
Collapse
|
21
|
Qi J, Li Q, Xin T, Lu Q, Lin J, Zhang Y, Luo H, Zhang F, Xing Y, Wang W, Cui D, Wang M. MCOLN1/TRPML1 in the lysosome: a promising target for autophagy modulation in diverse diseases. Autophagy 2024; 20:1712-1722. [PMID: 38522082 PMCID: PMC11262240 DOI: 10.1080/15548627.2024.2333715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024] Open
Abstract
MCOLN1/TRPML1 is a nonselective cationic channel specifically localized to the late endosome and lysosome. With its property of mediating the release of several divalent cations such as Ca2+, Zn2+ and Fe2+ from the lysosome to the cytosol, MCOLN1 plays a pivotal role in regulating a variety of cellular events including endocytosis, exocytosis, lysosomal biogenesis, lysosome reformation, and especially in Macroautophagy/autophagy. Autophagy is a highly conserved catabolic process that maintains cytoplasmic integrity by removing superfluous proteins and damaged organelles. Acting as the terminal compartments, lysosomes are crucial for the completion of the autophagy process. This review delves into the emerging role of MCOLN1 in controlling the autophagic process by regulating lysosomal ionic homeostasis, thereby governing the fundamental functions of lysosomes. Furthermore, this review summarizes the physiological relevance as well as molecular mechanisms through which MCOLN1 orchestrates autophagy, consequently influencing mitochondria turnover, cell apoptosis and migration. In addition, we have illustrated the implications of MCOLN1-regulated autophagy in the pathological process of cancer and myocardial ischemia-reperfusion (I/R) injury. In summary, given the involvement of MCOLN1-mediated autophagy in the pathogenesis of cancer and myocardial I/R injury, targeting MCOLN1 May provide clues for developing new therapeutic strategies for the treatment of these diseases. Exploring the regulation of MCOLN1-mediated autophagy in diverse diseases contexts will surely broaden our understanding of this pathway and offer its potential as a promising drug target.Abbreviation: CCCP:carbonyl cyanide3-chlorophenylhydrazone; CQ:chloroquine; HCQ: hydroxychloroquine;I/R: ischemia-reperfusion; MAP1LC3/LC3:microtubule associated protein 1 light chain 3; MCOLN1/TRPML1:mucolipin TRP cation channel 1; MLIV: mucolipidosis type IV; MTORC1:MTOR complex 1; ROS: reactive oxygenspecies; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Jiansong Qi
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qingqing Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tianli Xin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qixia Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jinyi Lin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Haiting Luo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Feifei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanhong Xing
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Derong Cui
- Department of Anesthesiology, The Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengmeng Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital, China of Medical University, Shenyang, LiaoningChina
| |
Collapse
|
22
|
Gao J, Li H, Lv H, Cheng X. Mutation of TRPML1 Channel and Pathogenesis of Neurodegeneration in Haimeria. Mol Neurobiol 2024; 61:4992-5001. [PMID: 38157120 DOI: 10.1007/s12035-023-03874-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Neurodegenerative diseases, a group of debilitating disorders, have garnered increasing attention due to their escalating prevalence, particularly among aging populations. Alzheimer's disease (AD) reigns as a prominent exemplar within this category, distinguished by its relentless progression of cognitive impairment and the accumulation of aberrant protein aggregates within the intricate landscape of the brain. While the intricate pathogenesis of neurodegenerative diseases has been the subject of extensive investigation, recent scientific inquiry has unveiled a novel player in this complex scenario-transient receptor potential mucolipin 1 (TRPML1) channels. This comprehensive review embarks on an exploration of the intricate interplay between TRPML1 channels and neurodegenerative diseases, with an explicit spotlight on Alzheimer's disease. It immerses itself in the intricate molecular mechanisms governing TRPML1 channel functionality and elucidates their profound implications for the well-being of neurons. Furthermore, the review ventures into the realm of therapeutic potential, pondering the possibilities and challenges associated with targeting TRPML1 channels as a promising avenue for the amelioration of neurodegenerative disorders. As we traverse this multifaceted terrain of neurodegeneration and the enigmatic role of TRPML1 channels, we embark on a journey that not only broadens our understanding of the intricate machinery governing neuronal health but also holds promise for the development of innovative therapeutic interventions in the relentless battle against neurodegenerative diseases.
Collapse
Affiliation(s)
- Junqing Gao
- Department of Neurology, Shaanxi Provincial People's Hospital, Shaanxi, Xi'an, 710068, China
| | - Huanhuan Li
- Department of Neurology, Tangdu Hospital, The Fourth Military Medical University, Shaanxi, Xi'an, 710038, China
| | - Hua Lv
- Department of Neurology, Shaanxi Provincial People's Hospital, Shaanxi, Xi'an, 710068, China
| | - Xiansong Cheng
- Department of Neurology, Shaanxi Provincial People's Hospital, Shaanxi, Xi'an, 710068, China.
| |
Collapse
|
23
|
Wu H, Fu M, Wu M, Cao Z, Zhang Q, Liu Z. Emerging mechanisms and promising approaches in pancreatic cancer metabolism. Cell Death Dis 2024; 15:553. [PMID: 39090116 PMCID: PMC11294586 DOI: 10.1038/s41419-024-06930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Pancreatic cancer is an aggressive cancer with a poor prognosis. Metabolic abnormalities are one of the hallmarks of pancreatic cancer, and pancreatic cancer cells can adapt to biosynthesis, energy intake, and redox needs through metabolic reprogramming to tolerate nutrient deficiency and hypoxic microenvironments. Pancreatic cancer cells can use glucose, amino acids, and lipids as energy to maintain malignant growth. Moreover, they also metabolically interact with cells in the tumour microenvironment to change cell fate, promote tumour progression, and even affect immune responses. Importantly, metabolic changes at the body level deserve more attention. Basic research and clinical trials based on targeted metabolic therapy or in combination with other treatments are in full swing. A more comprehensive and in-depth understanding of the metabolic regulation of pancreatic cancer cells will not only enrich the understanding of the mechanisms of disease progression but also provide inspiration for new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hao Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengdi Fu
- Department of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhen Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qiyao Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ziwen Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
24
|
Hu M, Feng X, Liu Q, Liu S, Huang F, Xu H. The ion channels of endomembranes. Physiol Rev 2024; 104:1335-1385. [PMID: 38451235 PMCID: PMC11381013 DOI: 10.1152/physrev.00025.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 02/20/2024] [Accepted: 02/25/2024] [Indexed: 03/08/2024] Open
Abstract
The endomembrane system consists of organellar membranes in the biosynthetic pathway [endoplasmic reticulum (ER), Golgi apparatus, and secretory vesicles] as well as those in the degradative pathway (early endosomes, macropinosomes, phagosomes, autophagosomes, late endosomes, and lysosomes). These endomembrane organelles/vesicles work together to synthesize, modify, package, transport, and degrade proteins, carbohydrates, and lipids, regulating the balance between cellular anabolism and catabolism. Large ion concentration gradients exist across endomembranes: Ca2+ gradients for most endomembrane organelles and H+ gradients for the acidic compartments. Ion (Na+, K+, H+, Ca2+, and Cl-) channels on the organellar membranes control ion flux in response to cellular cues, allowing rapid informational exchange between the cytosol and organelle lumen. Recent advances in organelle proteomics, organellar electrophysiology, and luminal and juxtaorganellar ion imaging have led to molecular identification and functional characterization of about two dozen endomembrane ion channels. For example, whereas IP3R1-3 channels mediate Ca2+ release from the ER in response to neurotransmitter and hormone stimulation, TRPML1-3 and TMEM175 channels mediate lysosomal Ca2+ and H+ release, respectively, in response to nutritional and trafficking cues. This review aims to summarize the current understanding of these endomembrane channels, with a focus on their subcellular localizations, ion permeation properties, gating mechanisms, cell biological functions, and disease relevance.
Collapse
Affiliation(s)
- Meiqin Hu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Xinghua Feng
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Qiang Liu
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Siyu Liu
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Fangqian Huang
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Haoxing Xu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
25
|
Jacquemyn J, Ralhan I, Ioannou MS. Driving factors of neuronal ferroptosis. Trends Cell Biol 2024; 34:535-546. [PMID: 38395733 DOI: 10.1016/j.tcb.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024]
Abstract
Ferroptosis is an oxidative form of iron-dependent cell death characterized by the accumulation of lipid peroxides on membranes. Iron and lipids containing polyunsaturated fatty acids are essential for this process. Ferroptosis is central to several neurological diseases and underlies the importance of balanced iron and polyunsaturated fatty acid metabolism in the brain, particularly in neurons. Here, we reflect on the potential links between neuronal physiology and the accumulation of iron and peroxidated lipids, the mechanisms neurons use to protect themselves from ferroptosis, and the relationship between pathogenic protein deposition and ferroptosis in neurodegenerative disease. We propose that the unique physiology of neurons makes them especially vulnerable to ferroptosis.
Collapse
Affiliation(s)
- Julie Jacquemyn
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada; Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Isha Ralhan
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada; Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Maria S Ioannou
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada; Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Cell Biology, University of Alberta, Edmonton, AB T6G 2R3, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
26
|
Gu J, Guo C, Ruan J, Li K, Zhou Y, Gong X, Shi H. From ferroptosis to cuproptosis, and calcicoptosis, to find more novel metals-mediated distinct form of regulated cell death. Apoptosis 2024; 29:586-604. [PMID: 38324163 DOI: 10.1007/s10495-023-01927-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 02/08/2024]
Abstract
Regulated cell death (RCD), also known as programmed cell death (PCD), plays a critical role in various biological processes, such as tissue injury/repair, development, and homeostasis. Dysregulation of RCD pathways can lead to the development of many human diseases, such as cancer, neurodegenerative disorders, and cardiovascular diseases. Maintaining proper metal ion homeostasis is critical for human health. However, imbalances in metal levels within cells can result in cytotoxicity and cell death, leading to a variety of diseases and health problems. In recent years, new types of metal overload-induced cell death have been identified, including ferroptosis, cuproptosis, and calcicoptosis. This has prompted us to examine the three defined metal-dependent cell death types, and discuss other metals-induced ferroptosis, cuproptosis, and disrupted Ca2+ homeostasis, as well as the roles of Zn2+ in metals' homeostasis and related RCD. We have reviewed the connection between metals-induced RCD and various diseases, as well as the underlying mechanisms. We believe that further research in this area will lead to the discovery of novel types of metal-dependent RCD, a better understanding of the underlying mechanisms, and the development of new therapeutic strategies for human diseases.
Collapse
Affiliation(s)
- Jie Gu
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| | - Chuanzhi Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| | - Jiacheng Ruan
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| | - Kongdong Li
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| | - Yang Zhou
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| | - Xun Gong
- Department of Rheumatology & Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212013, China.
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
27
|
Fan C, Wu H, Du X, Li C, Zeng W, Qu L, Cang C. Inhibition of lysosomal TRPML1 channel eliminates breast cancer stem cells by triggering ferroptosis. Cell Death Discov 2024; 10:256. [PMID: 38802335 PMCID: PMC11130215 DOI: 10.1038/s41420-024-02026-y] [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: 01/23/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Cancer stem cells (CSCs) are a sub-population of cells possessing high tumorigenic potential, which contribute to therapeutic resistance, metastasis and recurrence. Eradication of CSCs is widely recognized as a crucial factor in improving patient prognosis, yet the effective targeting of these cells remains a major challenge. Here, we show that the lysosomal cation channel TRPML1 represents a promising target for CSCs. TRPML1 is highly expressed in breast cancer cells and exhibits sensitivity to salinomycin, a drug known to selectively eliminate CSCs. Pharmacological inhibition and genetic depletion of TRPML1 promote ferroptosis in breast CSCs, reduce their stemness, and enhance the sensitivity of breast cancer cells to chemotherapy drug doxorubicin. The inhibition and knockout of TRPML1 also demonstrate significant suppression of tumor formation and growth in the mouse xenograft model. These findings suggest that targeting TRPML1 to eliminate CSCs may be an effective strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Chunhong Fan
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Haotian Wu
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Xin Du
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Canjun Li
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230061, Anhui, China
| | - Wenping Zeng
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Lili Qu
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| | - Chunlei Cang
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230061, Anhui, China.
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
28
|
Pan T, Gao Y, Xu G, Yu L, Xu Q, Yu J, Liu M, Zhang C, Ma Y, Li Y. Widespread transcriptomic alterations of transient receptor potential channel genes in cancer. Brief Funct Genomics 2024; 23:214-227. [PMID: 37288496 DOI: 10.1093/bfgp/elad023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/09/2023] Open
Abstract
Ion channels, in particular transient-receptor potential (TRP) channels, are essential genes that play important roles in many physiological processes. Emerging evidence has demonstrated that TRP genes are involved in a number of diseases, including various cancer types. However, we still lack knowledge about the expression alterations landscape of TRP genes across cancer types. In this review, we comprehensively reviewed and summarised the transcriptomes from more than 10 000 samples in 33 cancer types. We found that TRP genes were widespreadly transcriptomic dysregulated in cancer, which was associated with clinical survival of cancer patients. Perturbations of TRP genes were associated with a number of cancer pathways across cancer types. Moreover, we reviewed the functions of TRP family gene alterations in a number of diseases reported in recent studies. Taken together, our study comprehensively reviewed TRP genes with extensive transcriptomic alterations and their functions will directly contribute to cancer therapy and precision medicine.
Collapse
Affiliation(s)
- Tao Pan
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Yueying Gao
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Gang Xu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | | | - Qi Xu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Jinyang Yu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Meng Liu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Can Zhang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| | - Yongsheng Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Department of Reproductive Medicine, the First Affliated Hospital of Hainan Medical University, College of Biomedical Information and Engineering, Hainan Medical University, Haikou, Hainan 571199, China
| |
Collapse
|
29
|
Luan M, Feng Z, Zhu W, Xing Y, Ma X, Zhu J, Wang Y, Jia Y. Mechanism of metal ion-induced cell death in gastrointestinal cancer. Biomed Pharmacother 2024; 174:116574. [PMID: 38593706 DOI: 10.1016/j.biopha.2024.116574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/11/2024] Open
Abstract
Gastrointestinal (GI) cancer is one of the most severe types of cancer, with a significant impact on human health worldwide. Due to the urgent demand for more effective therapeutic strategies against GI cancers, novel research on metal ions for treating GI cancers has attracted increasing attention. Currently, with accumulating research on the relationship between metal ions and cancer therapy, several metal ions have been discovered to induce cell death. In particular, the three novel modes of cell death, including ferroptosis, cuproptosis, and calcicoptosis, have become focal points of research in the field of cancer. Meanwhile, other metal ions have also been found to trigger cell death through various mechanisms. Accordingly, this review focuses on the mechanisms of metal ion-induced cell death in GI cancers, hoping to provide theoretical support for further GI cancer therapies.
Collapse
Affiliation(s)
- Muhua Luan
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Zhaotian Feng
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China; Department of Medical Laboratory, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Wenshuai Zhu
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Jingyu Zhu
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yunshan Wang
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yanfei Jia
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China; Department of Medical Laboratory, Weifang Medical University, Weifang 261053, People's Republic of China.
| |
Collapse
|
30
|
Zhou QY, Ren C, Li JY, Wang L, Duan Y, Yao RQ, Tian YP, Yao YM. The crosstalk between mitochondrial quality control and metal-dependent cell death. Cell Death Dis 2024; 15:299. [PMID: 38678018 PMCID: PMC11055915 DOI: 10.1038/s41419-024-06691-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Mitochondria are the centers of energy and material metabolism, and they also serve as the storage and dispatch hubs of metal ions. Damage to mitochondrial structure and function can cause abnormal levels and distribution of metal ions, leading to cell dysfunction and even death. For a long time, mitochondrial quality control pathways such as mitochondrial dynamics and mitophagy have been considered to inhibit metal-induced cell death. However, with the discovery of new metal-dependent cell death including ferroptosis and cuproptosis, increasing evidence shows that there is a complex relationship between mitochondrial quality control and metal-dependent cell death. This article reviews the latest research results and mechanisms of crosstalk between mitochondrial quality control and metal-dependent cell death in recent years, as well as their involvement in neurodegenerative diseases, tumors and other diseases, in order to provide new ideas for the research and treatment of related diseases.
Collapse
Affiliation(s)
- Qi-Yuan Zhou
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Chao Ren
- Department of Pulmonary and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing-Yan Li
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lu Wang
- Department of Critical Care Medicine, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yu Duan
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou, 423000, China
| | - Ren-Qi Yao
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
- Medical Innovation Research Division, Translational Medicine Research Center and the Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| | - Ying-Ping Tian
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Yong-Ming Yao
- Medical Innovation Research Division, Translational Medicine Research Center and the Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
31
|
Radin DP, Shifman S, Outhwaite IR, Sharma A, Bases R, Seeliger MA, Tsirka SE. Lucanthone, a Potential PPT1 Inhibitor, Perturbs Stemness, Reduces Tumor Microtube Formation, and Slows the Growth of Temozolomide-Resistant Gliomas In Vivo. J Pharmacol Exp Ther 2024; 389:51-60. [PMID: 38296645 PMCID: PMC10949164 DOI: 10.1124/jpet.123.002021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/28/2023] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Glioblastoma (GBM) is the most frequently diagnosed primary central nervous system tumor in adults. Despite the standard of care therapy, which includes surgical resection, temozolomide chemotherapy, radiation and the newly added tumor-treating fields, median survival remains only ∼20 months. Unfortunately, GBM has a ∼100% recurrence rate, but after recurrence there are no Food and Drug Administration-approved therapies to limit tumor growth and enhance patient survival, as these tumors are resistant to temozolomide (TMZ). Recently, our laboratory reported that lucanthone slows GBM by inhibiting autophagic flux through lysosome targeting and decreases the number of Olig2+ glioma stem-like cells (GSC) in vitro and in vivo. We now additionally report that lucanthone efficiently abates stemness in patient-derived GSC and reduces tumor microtube formation in GSC, an emerging hallmark of treatment resistance in GBM. In glioma tumors derived from cells with acquired resistance to TMZ, lucanthone retains the ability to perturb tumor growth, inhibits autophagy by targeting lysosomes, and reduces Olig2 positivity. We also find that lucanthone may act as an inhibitor of palmitoyl protein thioesterase 1. Our results suggest that lucanthone may function as a potential treatment option for GBM tumors that are not amenable to TMZ treatment. SIGNIFICANCE STATEMENT: We report that the antischistosome agent lucanthone impedes tumor growth in a preclinical model of temozolomide-resistant glioblastoma and reduces the numbers of stem-like glioma cells. In addition, it acts as an autophagy inhibitor, and its mechanism of action may be via inhibition of palmitoyl protein thioesterase 1. As there are no defined therapies approved for recurrent, TMZ-resistant tumor, lucanthone could emerge as a treatment for glioblastoma tumors that may not be amenable to TMZ both in the newly diagnosed and recurrent settings.
Collapse
Affiliation(s)
- Daniel P Radin
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Sophie Shifman
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Ian R Outhwaite
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Aryan Sharma
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Robert Bases
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Markus A Seeliger
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Stella E Tsirka
- Department of Pharmacological Sciences (D.P.R., S.S., I.R.O., A.S., M.A.S., S.E.T.) and Stony Brook Medical Scientist Training Program (D.P.R., S.S., I.R.O.), Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| |
Collapse
|
32
|
Yang X, Li W, Ding M, Liu KJ, Qi Z, Zhao Y. Contribution of zinc accumulation to ischemic brain injury and its mechanisms about oxidative stress, inflammation, and autophagy: an update. Metallomics 2024; 16:mfae012. [PMID: 38419293 DOI: 10.1093/mtomcs/mfae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024]
Abstract
Ischemic stroke is a leading cause of death and disability worldwide, and presently, there is no effective neuroprotective therapy. Zinc is an essential trace element that plays important physiological roles in the central nervous system. Free zinc concentration is tightly regulated by zinc-related proteins in the brain under normal conditions. Disruption of zinc homeostasis, however, has been found to play an important role in the mechanism of brain injury following ischemic stroke. A large of free zinc releases from storage sites after cerebral ischemia, which affects the functions and survival of nerve cells, including neurons, astrocytes, and microglia, resulting in cell death. Ischemia-triggered intracellular zinc accumulation also disrupts the function of blood-brain barrier via increasing its permeability, impairing endothelial cell function, and altering tight junction levels. Oxidative stress and neuroinflammation have been reported to be as major pathological mechanisms in cerebral ischemia/reperfusion injury. Studies have showed that the accumulation of intracellular free zinc could impair mitochondrial function to result in oxidative stress, and form a positive feedback loop between zinc accumulation and reactive oxygen species production, which leads to a series of harmful reactions. Meanwhile, elevated intracellular zinc leads to neuroinflammation. Recent studies also showed that autophagy is one of the important mechanisms of zinc toxicity after ischemic injury. Interrupting the accumulation of zinc will reduce cerebral ischemia injury and improve neurological outcomes. This review summarizes the role of zinc toxicity in cellular and tissue damage following cerebral ischemia, focusing on the mechanisms about oxidative stress, inflammation, and autophagy.
Collapse
Affiliation(s)
- Xueqi Yang
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China
- Beijing Geriatric Medical Research Center, Beijing 100053, China
| | - Wei Li
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China
- Beijing Geriatric Medical Research Center, Beijing 100053, China
| | - Mao Ding
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China
| | - Ke Jian Liu
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Zhifeng Qi
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China
- Beijing Geriatric Medical Research Center, Beijing 100053, China
| | - Yongmei Zhao
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China
- Beijing Geriatric Medical Research Center, Beijing 100053, China
| |
Collapse
|
33
|
Li Y, Lv C, Li Z, Chen C, Cheng Y. Magnetic modulation of lysosomes for cancer therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1947. [PMID: 38488191 DOI: 10.1002/wnan.1947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 03/19/2024]
Abstract
Lysosomes play a central role in biochemical signal transduction and oxidative stress in cells. Inducing lysosome membrane penetration (LMP) to cause lysosomal-dependent cell death (LCD) in tumor cells is an effective strategy for cancer therapy. Chemical drugs can destroy the stability of lysosomes by neutralizing protons within the lysosomes or enhancing the fragility of the lysosomal membranes. However, there remain several unsolved problems of traditional drugs in LMP induction due to insufficient lysosomal targeting, fast metabolism, and toxicity in normal cells. With the development of nanotechnology, magnetic nanoparticles have been demonstrated to target lysosomes naturally, providing a versatile tool for lysosomal modulation. Combined with excellent tissue penetration and spatiotemporal manipulability of magnetic fields, magnetic modulation of lysosomes progresses rapidly in inducing LMP and LCD for cancer therapy. This review comprehensively discussed the strategies of magnetic modulation of lysosomes for cancer therapy. The intrinsic mechanisms of LMP-induced LCD were first introduced. Then, the modulation of lysosomes by diverse physical outputs of magnetic fields was emphatically discussed. Looking forward, this review will shed the light on the prospect of magnetic modulation of lysosomes, inspiring future research of magnetic modulation strategy in cancer therapy. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Yingze Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, School of Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cheng Lv
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, School of Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Zhenguang Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, School of Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Cheng
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, School of Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
34
|
Zhang Z, Liang X, Yang X, Liu Y, Zhou X, Li C. Advances in Nanodelivery Systems Based on Metabolism Reprogramming Strategies for Enhanced Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6689-6708. [PMID: 38302434 DOI: 10.1021/acsami.3c15686] [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: 02/03/2024]
Abstract
Tumor development and metastasis are closely related to the complexity of the metabolism network. Recently, metabolism reprogramming strategies have attracted much attention in tumor metabolism therapy. Although there is preliminary success of metabolism therapy agents, their therapeutic effects have been restricted by the effective reaching of the tumor sites of drugs. Nanodelivery systems with unique physical properties and elaborate designs can specifically deliver to the tumors. In this review, we first summarize the research progress of nanodelivery systems based on tumor metabolism reprogramming strategies to enhance therapies by depleting glucose, inhibiting glycolysis, depleting lactic acid, inhibiting lipid metabolism, depleting glutamine and glutathione, and disrupting metal metabolisms combined with other therapies, including chemotherapy, radiotherapy, photodynamic therapy, etc. We further discuss in detail the advantages of nanodelivery systems based on tumor metabolism reprogramming strategies for tumor therapy. As well as the opportunities and challenges for integrating nanodelivery systems into tumor metabolism therapy, we analyze the outlook for these emerging areas. This review is expected to improve our understanding of modulating tumor metabolisms for enhanced therapy.
Collapse
Affiliation(s)
- Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiangyu Zhou
- Department of Thyroid and Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
35
|
Pan Y, Zhao Q, He H, Qi Y, Bai Y, Zhao J, Yang Y. TRPML1 as a potential therapeutic target for triple-negative breast cancer: a review. Front Oncol 2023; 13:1326023. [PMID: 38156109 PMCID: PMC10753766 DOI: 10.3389/fonc.2023.1326023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 12/01/2023] [Indexed: 12/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most refractory subtype of breast cancer, and effective treatments are urgently needed owing to its poor prognosis. Surgery, radiotherapy, and chemotherapy, alone or in combination, are the leading choices for TNBC therapy. Although promising approaches and procedures have emerged, several challenges, such as off-target effects, drug resistance, and severe side effects, remain to be addressed. Recently, transient receptor potential channel mucolipin 1 (TRPML1) has attracted the attention of researchers because its expression has been implicated in numerous diseases, including cancer. TRPML1 regulates biological events and signaling pathways, including autophagic flux, exocytosis, ionic homeostasis, and lysosomal biogenesis, all contributing to tumorigenesis and cancer progression. TRPML1 also functions as a building block for cancer cell growth, mitogenic signaling, priming tissues for metastasis, and activation of transcriptional programs, processes involved in several malignant tumors. This review provides an overview of breast cancer epidemiology and diagnostic techniques and then discusses the existing therapeutics. Additionally, we elaborate on the development of, and associated challenges to, TNBC diagnostics and treatment and the feasibility of TRPML1 as a therapeutic target for TNBC.
Collapse
Affiliation(s)
- Ying Pan
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Qiancheng Zhao
- Department of Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Haitao He
- Department of Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yubo Qi
- First Hospital of Jilin University, Changchun, Jilin, China
| | - Yujie Bai
- First Hospital of Jilin University, Changchun, Jilin, China
| | - Jia Zhao
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yiming Yang
- Department of Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
36
|
Deng Z, Chen S, Liu G, Zhu G. Unlocking the potential of platinum drugs: organelle-targeted small-molecule platinum complexes for improved anticancer performance. RSC Chem Biol 2023; 4:1003-1013. [PMID: 38033725 PMCID: PMC10685827 DOI: 10.1039/d3cb00087g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/29/2023] [Indexed: 12/02/2023] Open
Abstract
Platinum-based drugs have revolutionized cancer chemotherapy; however, their therapeutic efficacy has been limited by severe side effects and drug resistance. Recently, approaches that target specific organelles in cancer cells have emerged as attractive alternatives to overcome these challenges. Many studies have validated these strategies and highlighted that organelle-targeted platinum complexes demonstrate increased anticancer activity, the ability to overcome drug resistance, novel molecular mechanisms, or even lower toxicity. This review provides a brief summary of various organelle-targeting strategies that promote the accumulation of platinum complexes in certain intracellular areas, such as the nucleus, mitochondria, endoplasmic reticulum (ER), and lysosomes. Moreover, the mechanisms through which these strategies improve anticancer performance, overcome drug resistance, and alter the action mode of conventional platinum drugs are discussed. By providing an extensive account of platinum complexes targeting different organelles, this review aims to assist researchers in understanding the design principles, identifying potential targets, and fostering innovative ideas for the development of platinum complexes.
Collapse
Affiliation(s)
- Zhiqin Deng
- Department of Chemistry, City University of Hong Kong Hong Kong SAR P. R. China
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 P. R. China
- School of Medicine, Chongqing University Chongqing 400030 P. R. China
| | - Shu Chen
- Department of Chemistry, City University of Hong Kong Hong Kong SAR P. R. China
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 P. R. China
| | - Gongyuan Liu
- Department of Chemistry, City University of Hong Kong Hong Kong SAR P. R. China
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 P. R. China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong Hong Kong SAR P. R. China
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 P. R. China
| |
Collapse
|
37
|
Chen Q, Wu J, Li X, Ye Z, Yang H, Mu L. Amphibian-Derived Natural Anticancer Peptides and Proteins: Mechanism of Action, Application Strategies, and Prospects. Int J Mol Sci 2023; 24:13985. [PMID: 37762285 PMCID: PMC10530844 DOI: 10.3390/ijms241813985] [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: 07/18/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 09/29/2023] Open
Abstract
Cancer is one of the major diseases that seriously threaten human life. Traditional anticancer therapies have achieved remarkable efficacy but have also some unavoidable side effects. Therefore, more and more research focuses on highly effective and less-toxic anticancer substances of natural origin. Amphibian skin is rich in active substances such as biogenic amines, alkaloids, alcohols, esters, peptides, and proteins, which play a role in various aspects such as anti-inflammatory, immunomodulatory, and anticancer functions, and are one of the critical sources of anticancer substances. Currently, a range of natural anticancer substances are known from various amphibians. This paper aims to review the physicochemical properties, anticancer mechanisms, and potential applications of these peptides and proteins to advance the identification and therapeutic use of natural anticancer agents.
Collapse
Affiliation(s)
| | | | | | | | - Hailong Yang
- Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Lixian Mu
- Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
38
|
Hadian K, Stockwell BR. The therapeutic potential of targeting regulated non-apoptotic cell death. Nat Rev Drug Discov 2023; 22:723-742. [PMID: 37550363 DOI: 10.1038/s41573-023-00749-8] [Citation(s) in RCA: 148] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 08/09/2023]
Abstract
Cell death is critical for the development and homeostasis of almost all multicellular organisms. Moreover, its dysregulation leads to diverse disease states. Historically, apoptosis was thought to be the major regulated cell death pathway, whereas necrosis was considered to be an unregulated form of cell death. However, research in recent decades has uncovered several forms of regulated necrosis that are implicated in degenerative diseases, inflammatory conditions and cancer. The growing insight into these regulated, non-apoptotic cell death pathways has opened new avenues for therapeutic targeting. Here, we describe the regulatory pathways of necroptosis, pyroptosis, parthanatos, ferroptosis, cuproptosis, lysozincrosis and disulfidptosis. We discuss small-molecule inhibitors of the pathways and prospects for future drug discovery. Together, the complex mechanisms governing these pathways offer strategies to develop therapeutics that control non-apoptotic cell death.
Collapse
Affiliation(s)
- Kamyar Hadian
- Research Unit Signaling and Translation, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Brent R Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA.
| |
Collapse
|
39
|
Minckley TF, Salvagio LA, Fudge DH, Verhey K, Markus SM, Qin Y. Zn2+ decoration of microtubules arrests axonal transport and displaces tau, doublecortin, and MAP2C. J Cell Biol 2023; 222:e202208121. [PMID: 37326602 PMCID: PMC10276529 DOI: 10.1083/jcb.202208121] [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: 08/22/2022] [Revised: 03/31/2023] [Accepted: 05/11/2023] [Indexed: 06/17/2023] Open
Abstract
Intracellular Zn2+ concentrations increase via depolarization-mediated influx or intracellular release, but the immediate effects of Zn2+ signals on neuron function are not fully understood. By simultaneous recording of cytosolic Zn2+ and organelle motility, we find that elevated Zn2+ (IC50 ≈ 5-10 nM) reduces both lysosomal and mitochondrial motility in primary rat hippocampal neurons and HeLa cells. Using live-cell confocal microscopy and in vitro single-molecule TIRF imaging, we reveal that Zn2+ inhibits activity of motor proteins (kinesin and dynein) without disrupting their microtubule binding. Instead, Zn2+ directly binds to microtubules and selectively promotes detachment of tau, DCX, and MAP2C, but not MAP1B, MAP4, MAP7, MAP9, or p150glued. Bioinformatic predictions and structural modeling show that the Zn2+ binding sites on microtubules partially overlap with the microtubule binding sites of tau, DCX, dynein, and kinesin. Our results reveal that intraneuronal Zn2+ regulates axonal transport and microtubule-based processes by interacting with microtubules.
Collapse
Affiliation(s)
- Taylor F. Minckley
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | | | - Dylan H. Fudge
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Kristen Verhey
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Steven M. Markus
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Yan Qin
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| |
Collapse
|
40
|
Dorward AM, Stewart AJ, Pitt SJ. The role of Zn2+ in shaping intracellular Ca2+ dynamics in the heart. J Gen Physiol 2023; 155:e202213206. [PMID: 37326614 PMCID: PMC10276528 DOI: 10.1085/jgp.202213206] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/18/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Increasing evidence suggests that Zn2+ acts as a second messenger capable of transducing extracellular stimuli into intracellular signaling events. The importance of Zn2+ as a signaling molecule in cardiovascular functioning is gaining traction. In the heart, Zn2+ plays important roles in excitation-contraction (EC) coupling, excitation-transcription coupling, and cardiac ventricular morphogenesis. Zn2+ homeostasis in cardiac tissue is tightly regulated through the action of a combination of transporters, buffers, and sensors. Zn2+ mishandling is a common feature of various cardiovascular diseases. However, the precise mechanisms controlling the intracellular distribution of Zn2+ and its variations during normal cardiac function and during pathological conditions are not fully understood. In this review, we consider the major pathways by which the concentration of intracellular Zn2+ is regulated in the heart, the role of Zn2+ in EC coupling, and discuss how Zn2+ dyshomeostasis resulting from altered expression levels and efficacy of Zn2+ regulatory proteins are key drivers in the progression of cardiac dysfunction.
Collapse
Affiliation(s)
- Amy M. Dorward
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Alan J. Stewart
- School of Medicine, University of St Andrews, St Andrews, UK
| | | |
Collapse
|
41
|
Oh SJ, Hwang Y, Hur KY, Lee MS. Lysosomal Ca 2+ as a mediator of palmitate-induced lipotoxicity. Cell Death Discov 2023; 9:100. [PMID: 36944629 PMCID: PMC10030853 DOI: 10.1038/s41420-023-01379-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/23/2023] Open
Abstract
While the mechanism of lipotoxicity by palmitic acid (PA), an effector of metabolic stress in vitro and in vivo, has been extensively investigated, molecular details of lipotoxicity are still not fully characterized. Since recent studies reported that PA can exert lysosomal stress in addition to well-known ER and mitochondrial stress, we studied the role of lysosomal events in lipotoxicity by PA, focusing on lysosomal Ca2+. We found that PA induced accumulation of mitochondrial ROS and that mitochondrial ROS induced release of lysosomal Ca2+ due to lysosomal Ca2+ exit channel activation. Lysosomal Ca2+ release led to increased cytosolic Ca2+ which induced mitochondrial permeability transition (mPT). Chelation of cytoplasmic Ca2+ or blockade of mPT with olesoxime or decylubiquinone (DUB) suppressed lipotoxicity. Lysosomal Ca2+ release led to reduced lysosomal Ca2+ content which was replenished by ER Ca2+, the largest intracellular Ca2+ reservoir (ER → lysosome Ca2+ refilling), which in turn activated store-operated Ca2+ entry (SOCE). Inhibition of ER → lysosome Ca2+ refilling by blockade of ER Ca2+ exit channel using dantrolene or inhibition of SOCE using BTP2 inhibited lipotoxicity in vitro. Dantrolene or DUB also inhibited lipotoxic death of hepatocytes in vivo induced by administration of ethyl palmitate together with LPS. These results suggest a novel pathway of lipotoxicity characterized by mPT due to lysosomal Ca2+ release which was supplemented by ER → lysosome Ca2+ refilling and subsequent SOCE, and also suggest the potential role of modulation of ER → lysosome Ca2+ refilling by dantrolene or other blockers of ER Ca2+ exit channels in disease conditions characterized by lipotoxicity such as metabolic syndrome, diabetes, cardiomyopathy or nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Soo-Jin Oh
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Korea
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang Medical Center, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Yeseong Hwang
- Severance Biomedical Science Institute, Graduate school of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Kyu Yeon Hur
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung-Shik Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang Medical Center, Soonchunhyang University College of Medicine, Cheonan, Korea.
- Severance Biomedical Science Institute, Graduate school of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea.
| |
Collapse
|
42
|
Guan X, Yang J, Wang W, Zhao B, Hu S, Yu D, Yuan L, Shi Y, Xu J, Dong J, Wang J, Cheng XD, Qin JJ. Dual inhibition of MYC and SLC39A10 by a novel natural product STAT3 inhibitor derived from Chaetomium globosum suppresses tumor growth and metastasis in gastric cancer. Pharmacol Res 2023; 189:106703. [PMID: 36804016 DOI: 10.1016/j.phrs.2023.106703] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/06/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Gastric cancer remains one of the most common deadly diseases and lacks effective targeted therapies. In the present study, we confirmed that the signal transducer and activator of transcription 3 (STAT3) is highly expressed and associated with a poor prognosis in gastric cancer. We further identified a novel natural product inhibitor of STAT3, termed XYA-2, which interacts specifically with the SH2 domain of STAT3 (Kd= 3.29 μM) and inhibits IL-6-induced STAT3 phosphorylation at Tyr705 and nuclear translocation. XYA-2 inhibited the viability of seven human gastric cancer cell lines with 72-h IC50 values ranging from 0.5 to 0.7 μΜ. XYA-2 at 1 μΜ inhibited the colony formation and migration ability of MGC803 (72.6% and 67.6%, respectively) and MKN28 (78.5% and 96.6%, respectively) cells. In the in vivo studies, intraperitoneal administration of XYA-2 (10 mg/kg/day, 7 days/week) significantly suppressed 59.8% and 88.8% tumor growth in the MKN28-derived xenograft mouse model and MGC803-derived orthotopic mouse model, respectively. Similar results were obtained in a patient-derived xenograft (PDX) mouse model. Moreover, XYA-2 treatment extended the survival of mice bearing PDX tumors. The molecular mechanism studies based on transcriptomics and proteomics analyses indicated that XYA-2 might exert its anticancer activity by synergistically inhibiting the expression of MYC and SLC39A10, two downstream genes of STAT3 in vitro and in vivo. Together, these findings suggested that XYA-2 may be a potent STAT3 inhibitor for treating gastric cancer, and dual inhibition of MYC and SLC39A10 may be an effective therapeutic strategy for STAT3-activated cancer.
Collapse
Affiliation(s)
- Xiaoqing Guan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jing Yang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Weiyi Wang
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian, China.
| | - Bing Zhao
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Shiyu Hu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Dehua Yu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Li Yuan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yunfu Shi
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jingli Xu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jinyun Dong
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jinxin Wang
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xiang-Dong Cheng
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Jiang-Jiang Qin
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
43
|
Lomphithak T, Fadeel B. Die hard: cell death mechanisms and their implications in nanotoxicology. Toxicol Sci 2023; 192:kfad008. [PMID: 36752525 PMCID: PMC10109533 DOI: 10.1093/toxsci/kfad008] [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] [Indexed: 02/09/2023] Open
Abstract
Cell death is a fundamental biological process, and its fine-tuned regulation is required for life. However, the complexity of regulated cell death is often reduced to a matter of live-dead discrimination. Here, we provide a perspective on programmed or regulated cell death, focusing on apoptosis, pyroptosis, necroptosis, and ferroptosis (the latter three cell death modalities are examples of regulated necrosis). We also touch on other, recently described manifestations of (pathological) cell death including cuproptosis. Furthermore, we address how engineered nanomaterials impact on regulated cell death. We posit that an improved understanding of nanomaterial-induced perturbations of cell death may allow for a better prediction of the consequences of human exposure and could also yield novel approaches by which to mitigate these effects. Finally, we provide examples of the harnessing of nanomaterials to achieve cancer cell killing through the induction of regulated cell death.
Collapse
Affiliation(s)
- Thanpisit Lomphithak
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Bengt Fadeel
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
44
|
Kim B, Kim G, Jeon S, Cho WS, Jeon HP, Jung J. Zinc oxide nanoparticles trigger autophagy-mediated cell death through activating lysosomal TRPML1 in normal kidney cells. Toxicol Rep 2023; 10:529-536. [PMID: 37152410 PMCID: PMC10160241 DOI: 10.1016/j.toxrep.2023.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/09/2023] Open
Abstract
Zinc oxide nanoparticles (ZnO NPs) have been widely used in various materials including sunscreens, cosmetics, over-the-counter topical skin products, and pigments. As traces of the used ZnO NPs have been found in the kidney, it is crucial to uncover their potential risks. The aim of this study is to elucidate detrimental effects of ZnO NPs and the molecular mechanism behind their renal toxicity. Cytotoxic effects were measured by MTT assay after HK2 cells were exposed to ZnO NPs for 24 h and IC50 value was determined. ROS and intracellular Zn2+ levels were detected by flow cytometry, and localization of Zn2+ and lysosome was determined by confocal microscopy. Occurrence of autophagy and detection of autophagic flux were determined by Western blot and confocal microscopy, respectively. We performed unpaired student t test for two groups, and one-way ANOVA with Tukey's post hoc for over three groups. ZnO NPs induced cell death in human renal proximal tubule epithelial cells, HK2. Cytosolic Zn2+ caused autophagy-mediated cell death rather than apoptosis. Cytosolic Zn2+ processed in lysosome was released by TRPML1, and inhibition of TRPML1 significantly decreased autophagic flux and cell death. The findings of this study suggest that ZnO NPs strongly induce autophagy-mediated cell death in human kidney cells. Controlling TRPML1 can be potentially used to prevent the kidney from ZnO NPs-induced toxicity.
Collapse
Affiliation(s)
- Boyun Kim
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan, the Republic of Korea
| | - Gaeun Kim
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan, the Republic of Korea
- Graduate School of Chemical Safety Management, Kyungsung University, Busan, the Republic of Korea
| | - Soyeon Jeon
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, the Republic of Korea
| | - Wan-Seob Cho
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, the Republic of Korea
| | - Hyun Pyo Jeon
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan, the Republic of Korea
- Graduate School of Chemical Safety Management, Kyungsung University, Busan, the Republic of Korea
- Correspondence to: Department of SmartBio, College of Life and Health Science, Kyungsung University, 309 Suyeong-ro Room 507-2, Nam-gu, Busan 48434, the Republic of Korea.
| | - Jewon Jung
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan, the Republic of Korea
- Graduate School of Chemical Safety Management, Kyungsung University, Busan, the Republic of Korea
- Correspondence to: Department of SmartBio, College of Life and Health Science, Kyungsung University, 309 Suyeong-ro Room 507-2, Nam-gu, Busan 48434, the Republic of Korea.
| |
Collapse
|
45
|
Shen M, Fan X, Shen Y, Wang X, Wu R, Wang Y, Huang C, Zhao S, Zheng Y, Men R, Luo X, Yang L. Myeloid-derived suppressor cells ameliorate liver mitochondrial damage to protect against autoimmune hepatitis by releasing small extracellular vesicles. Int Immunopharmacol 2023; 114:109540. [PMID: 36516541 DOI: 10.1016/j.intimp.2022.109540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Autoimmune hepatitis (AIH) is an inflammatory liver disease that is associated with impaired self-tolerance. Myeloid-derived supprfessor cells (MDSCs) have been considered to exert counterregulatory effects on AIH. However, the specific mechanism underlying these effects is unclear. Herein, we investigated the efficacy and safety of MDSCs in protecting against AIH and explored the underlying mechanism. METHODS Circulating and liver MDSC expression levels in 71 AIH patients and 47 healthy control (HC) individuals were detected by flow cytometry and immunohistochemistry. The adoptive transfer of induced bone marrow-derived MDSCs (BM MDSCs) to AIH mice was used to explore the function of MDSCs. Hepatic injury and mitochondrial damage were evaluated by transaminase levels, histopathology, immunohistochemistry, transmission electron microscopy and western blotting. MDSCs were pretreated with the small extracellular vesicle (sEV) generation inhibitor GW4869 to explore the mechanism. Importantly, sEVs derived from MDSCs and MDSCs-GW4869 were injected into model mice to monitor mitochondrial function and biogenesis. RESULTS Circulating and liver MDSCs were expanded in AIH patients and mouse model. Furthermore, the follow-up data of AIH patients showed that immunosuppressive therapy further promoted the expansion of MDSCs. More importantly, the adoptive transfer of BM MDSCs to AIH mice effectively ameliorated liver injury and regulated the imbalance of the immune microenvironment. Additionally, BM MDSCs reduced liver mitochondrial damage and improved mitochondrial biogenesis. Mechanistically, sEVs derived from BM MDSCs showed the same biological effects as cells, and blocking sEV production weakened the function of BM MDSCs. Finally, multiple long-term administrations of BM MDSCs were proven to be safe in general. CONCLUSION In conclusion, MDSCs ameliorate liver mitochondrial damage to protect against autoimmune hepatitis by releasing small extracellular vesicles.
Collapse
Affiliation(s)
- Mengyi Shen
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoli Fan
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Shen
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoze Wang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruiqi Wu
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Wang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chen Huang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shenglan Zhao
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanyi Zheng
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruoting Men
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuefeng Luo
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Li Yang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
46
|
Zhang K, Yang W, Zhang Z, Ma K, Li L, Xu Y, Qiu J, Yu C, Zhou J, Cai L, Gong Y, Gong K. A Novel Cuproptosis-Related Prognostic Model and the Hub Gene FDX1 Predict the Prognosis and Correlate with Immune Infiltration in Clear Cell Renal Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:2124088. [PMID: 36536785 PMCID: PMC9759391 DOI: 10.1155/2022/2124088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 09/29/2023]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common malignancy of the urological system with poor prognosis. Cuproptosis is a recently discovered novel manner of cell death, and the hub gene FDX1 could promote cuproptosis. However, the potential roles of cuproptosis-related genes (CRGs) and FDX1 for predicting prognosis, the immune microenvironment, and therapeutic response have been poorly studied in ccRCC. In the present study, The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) data were downloaded. CRGs were subjected to prognosis analysis, and three of them were used to construct the prognostic model by least absolute shrinkage and selection operator (LASSO) regression. The CRGs prognostic model showed excellent performance. Moreover, based on the risk score of the model, the nomogram was developed to predict 1-year, 3-year, and 5-year survival. Furthermore, the hub gene of cuproptosis, FDX1, was an independent prognostic biomarker in multivariate Cox regression analysis. The pan-cancer analysis showed that FDX1 was significantly downregulated and closely related to prognosis in ccRCC among 33 cancer types. Lower FDX1 was also correlated with worse clinicopathologic features. The lower expression of FDX1 in ccRCC was verified in the external database and our own database, which may be caused by DNA methylation. We further demonstrated that the tumor mutational burden (TMB) and immune cell infiltration were related to the expression of FDX1. Immune response and drug sensitivity analysis revealed that immunotherapy or elesclomol may have a favorable treatment effect in the high FDX1 expression group and sunitinib or axitinib may work better in the low FDX1 expression group. In conclusion, we constructed a CRGs prognostic model and revealed that FDX1 could serve as a prognostic biomarker and predict therapeutic response in ccRCC. The study will provide a novel, precise, and individual treatment strategy for ccRCC patients.
Collapse
Affiliation(s)
- Kenan Zhang
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Wuping Yang
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Zedan Zhang
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Kaifang Ma
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Lei Li
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Yawei Xu
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Jianhui Qiu
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Chaojian Yu
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Jingcheng Zhou
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Lin Cai
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
47
|
Lin H, Wu D, Xiao J. Identification of key cuproptosis-related genes and their targets in patients with IgAN. BMC Nephrol 2022; 23:354. [PMID: 36329405 PMCID: PMC9635123 DOI: 10.1186/s12882-022-02991-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/25/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND IgA nephropathy (IgAN) is one of the most common forms of chronic glomerulonephritis, but the aetiology and pathogenesis remain unclear. Cuproptosis is a newly identified form of cell death that plays an important role in many diseases. Researchers have not clearly determined whether the expression of cuproptosis-related genes (CRGs) is involved in the pathogenesis of IgAN. METHODS The GSE93798, GSE50469 and GSE37460 datasets containing microarray data from patients with IgAN (63) and healthy controls (31) were downloaded from the GEO database. Immune cells and immune-related functions were analysed in patients with IgAN and controls, and genes were identified that may be related to cuproptosis. A logistic regression model was established according to the results, and then GO and KEGG enrichment analyses were performed. Finally, possible drugs were selected using the DSigDB database. RESULTS The subjects in the different groups showed significantly different fractions of immune cells and immune-related functions, and 11 genes related to cuproptosis may be involved in these processes. Based on these 11 genes, the ROC curve was plotted, and the AUC value was calculated (0.898, 95% CI: 0.839-0.958). The result revealed good predictability. Then, genes with P < 0.05 (lipoyltransferase 1, LIPT1) were selected to plot an ROC curve, and the AUC value was calculated (0.729, 95% CI: 0.636-0.821). Enrichment analyses showed that the TCA cycle and multiple metabolic pathways may also be involved in the occurrence of IgAN. Finally, 293 potential drugs that may be used to treat IgAN were identified based on these genes. CONCLUSION In this study, we identified some novel CRGs that may be involved in IgAN, among which LIPT1 was significantly differentially expressed. It may predict the risk of IgAN and provides a possible target for the treatment of IgAN. Further experimental studies are needed to explore how these CRGs mediate the occurrence and development of IgAN.
Collapse
Affiliation(s)
- Huagang Lin
- grid.413597.d0000 0004 1757 8802Department of Nephrology, Huadong Hospital Affiliated to Fudan University, 200040 Shanghai, P.R. China ,grid.8547.e0000 0001 0125 2443Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, P.R. China
| | - Deping Wu
- grid.413597.d0000 0004 1757 8802Department of Nephrology, Huadong Hospital Affiliated to Fudan University, 200040 Shanghai, P.R. China ,grid.8547.e0000 0001 0125 2443Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, P.R. China
| | - Jing Xiao
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, 200040, Shanghai, P.R. China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, P.R. China.
| |
Collapse
|
48
|
Immanuel T, Li J, Green TN, Bogdanova A, Kalev-Zylinska ML. Deregulated calcium signaling in blood cancer: Underlying mechanisms and therapeutic potential. Front Oncol 2022; 12:1010506. [PMID: 36330491 PMCID: PMC9623116 DOI: 10.3389/fonc.2022.1010506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/21/2022] [Indexed: 02/05/2023] Open
Abstract
Intracellular calcium signaling regulates diverse physiological and pathological processes. In solid tumors, changes to calcium channels and effectors via mutations or changes in expression affect all cancer hallmarks. Such changes often disrupt transport of calcium ions (Ca2+) in the endoplasmic reticulum (ER) or mitochondria, impacting apoptosis. Evidence rapidly accumulates that this is similar in blood cancer. Principles of intracellular Ca2+ signaling are outlined in the introduction. We describe different Ca2+-toolkit components and summarize the unique relationship between extracellular Ca2+ in the endosteal niche and hematopoietic stem cells. The foundational data on Ca2+ homeostasis in red blood cells is discussed, with the demonstration of changes in red blood cell disorders. This leads to the role of Ca2+ in neoplastic erythropoiesis. Then we expand onto the neoplastic impact of deregulated plasma membrane Ca2+ channels, ER Ca2+ channels, Ca2+ pumps and exchangers, as well as Ca2+ sensor and effector proteins across all types of hematologic neoplasms. This includes an overview of genetic variants in the Ca2+-toolkit encoding genes in lymphoid and myeloid cancers as recorded in publically available cancer databases. The data we compiled demonstrate that multiple Ca2+ homeostatic mechanisms and Ca2+ responsive pathways are altered in hematologic cancers. Some of these alterations may have genetic basis but this requires further investigation. Most changes in the Ca2+-toolkit do not appear to define/associate with specific disease entities but may influence disease grade, prognosis, treatment response, and certain complications. Further elucidation of the underlying mechanisms may lead to novel treatments, with the aim to tailor drugs to different patterns of deregulation. To our knowledge this is the first review of its type in the published literature. We hope that the evidence we compiled increases awareness of the calcium signaling deregulation in hematologic neoplasms and triggers more clinical studies to help advance this field.
Collapse
Affiliation(s)
- Tracey Immanuel
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Jixia Li
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan City, China
| | - Taryn N. Green
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Anna Bogdanova
- Red Blood Cell Research Group, Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| | - Maggie L. Kalev-Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Haematology Laboratory, Department of Pathology and Laboratory Medicine, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
49
|
Liu J, Lu Y, Dai Y, Shen Y, Zeng C, Liu X, Yu H, Deng J, Lu W. A comprehensive analysis and validation of cuproptosis-associated genes across cancers: Overall survival, the tumor microenvironment, stemness scores, and drug sensitivity. Front Genet 2022; 13:939956. [PMID: 36105090 PMCID: PMC9465292 DOI: 10.3389/fgene.2022.939956] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Cuproptosis is a novel type of cell death induced by copper. Cuproptosis-associated genes play a crucial part in oncogenesis and the growth and metastasis of tumors. However, the correlations among cuproptosis-associated genes, overall survival, the tumor microenvironment, and drug sensitivity remain unclear. Therefore, we performed an analysis of cuproptosis-associated genes across cancers.Methods: We downloaded RNA sequence expression data, clinical and survival data, stemness score data, and immune subtype data of cuproptosis-associated genes from the UCSC Xena. Next, we conducted differential analysis, expression analysis and correlation analysis across cancers with various R packages. Moreover, survival analysis and Cox hazard analysis were conducted to investigate the relationships between cuproptosis-associated genes and survival outcomes in various cancer types. Finally, we also analyzed the relationship among the levels of cuproptosis-associated genes across cancers, immune types, the tumor microenvironment, stemness scores, and drug sensitivity. Expression validation of cuproptosis-associated genes in renal cancer and normal tissues by immunohistochemical staining.Results: We found that 10 cuproptosis-associated genes (FDX1, LIAS, LIPT1, DLD, DLAT, PDHA1, PDHB, MTF1, GLS, and CDKN2A) were differently expressed in 18 tumors and normal tissues. Survival outcomes showed that cuproptosis-associated genes had prognostic value in various cancer types. Moreover, we identified that cuproptosis-associated genes had different levels in six immune subtypes. The study also indicated that the levels of most cuproptosis-associated genes were positively correlated with the RNAss and DNAss. FDX1, LIAS, LIPT1, DLD, DLAT, PDHA1, and PDHB were negatively correlated with immune scores and ESTIMATE scores. In addition, we identified the top 16 drugs strongly sensitivity to cuproptosis-associated genes according to the correlation coefficient. Finally, we also found that cuproptosis-associated genes were significantly correlated with immune subtype, clinical features, the tumor microenvironment, and drug sensitivity in Kidney renal clear cell carcinoma. And the results of immunohistochemical staining analysis was very consistent with the previous analysis.Conclusion: We performed an overall analysis to uncover the roles of cuproptosis-associated genes in differential expression, survival outcomes, immune subtypes, the tumor microenvironment, stemness scores, and cancer drug sensitivity across cancers.
Collapse
Affiliation(s)
- Jinsong Liu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu, China
| | - Yueyao Lu
- Department of Oncology, The Changzhou Clinical School of Nanjing Medical University, Jiangsu, China
| | - Yuyang Dai
- Department of Radiology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
| | - Ying Shen
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu, China
| | - Cheng Zeng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu, China
| | - Xiuling Liu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu, China
| | - Huayi Yu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu, China
| | - Jianzhong Deng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu, China
| | - Wenbin Lu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Jiangsu, China
- Department of Oncology, The Changzhou Clinical School of Nanjing Medical University, Jiangsu, China
- Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Jiangsu, China
- *Correspondence: Wenbin Lu,
| |
Collapse
|
50
|
Gu M, Hu M, Minckley T, Pinchi P, Xu H, Qin Y, Du W. A protocol to measure lysosomal Zn 2+ release through a genetically encoded Zn 2+ indicator. STAR Protoc 2022; 3:101453. [PMID: 35707683 PMCID: PMC9189625 DOI: 10.1016/j.xpro.2022.101453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Intracellular vesicles such as lysosomes contain micromolar to millimolar concentrations of Zn2+, and disturbing lysosomal Zn2+ homeostasis via lysosomal Zn2+ release leads to mitochondria damage and consequent lytic cell death. Methods have been developed to image cellular Zn2+ dynamics. Here, we present a protocol using GZnP3, a genetically encoded fluorescent Zn2+ indicator, to assess lysosomal Zn2+ release in cultured cells by fluorescence microscopy imaging. For complete details on the use and execution of this protocol, please refer to Du et al. (2021) or Minckley et al. (2019).
Collapse
Affiliation(s)
- Mingxue Gu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4104 Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dun Neurological Research Institute, Suite 1125, 1250 Mursund Street, Houston, TX 77030, USA
| | - Meiqin Hu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4104 Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109, USA
| | - Taylor Minckley
- Department of Biological Sciences, University of Denver, 2190 E. Iliff Avenue, Denver, CO 80208, USA
| | - Prateeksunder Pinchi
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4104 Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109, USA
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4104 Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109, USA
| | - Yan Qin
- Department of Biological Sciences, University of Denver, 2190 E. Iliff Avenue, Denver, CO 80208, USA
| | - Wanlu Du
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4104 Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109, USA
| |
Collapse
|