1
|
Sun B, Gao W, Yu X, Zhang C, Du H, Luo Y, Zhu J, Yang P, Zhang M. Charge regulated pH/NIR dual responsive nanoplatforms centered on cuproptosis for enhanced cancer theranostics. Biomaterials 2025; 315:122907. [PMID: 39476451 DOI: 10.1016/j.biomaterials.2024.122907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/23/2024] [Accepted: 10/20/2024] [Indexed: 12/09/2024]
Abstract
Multifunctional nanoplatforms capable of simultaneously executing multimodal therapy and imaging functions are of great potentials for cancer theranostics. We present an elegantly designed, easy-to-fabricate poly(acrylic acid)/mesoporous calcium phosphate/mesoporous copper phosphate nanosphere (PAA/mCaP/mCuP NS) with outstanding pH/NIR-sensitive multimodal-synergic anti-tumor effects. Optimal porous PAA NS scaffolds were prepared at room temperature by balancing the intra-PAA polymer and polymer-solvents Lennard-Jones potentials in a water:isopropyl alcohol (IPA) mix-solvent. Subsequent sponging of Ca2+ and Cu2+, and adsorption of PO43- to the PAA template were achieved through exquisite electrostatic interactions among ions and the ionizable PAA side-chain in an aqueous environment. This forms the basis for the tumor microenvironment pH-triggered release of Cu2+ to induce cuproptosis, as well as the photothermal effect originating from CuP, while Ca2+ can enhance the nanoplatform's biocompatibility and can damage mitochondria when overloaded. Lastly, PAA/mCaP/mCuP NSs still exhibit high drug loading efficiency for doxorubicin (DOX), enabling chemotherapy. Satisfactory anti-tumor effects of these modalities, along with their synergistic effects, were verified both in vitro and in vivo, with the NSs demonstrating good biodegradation in the latter. The fabricated NS itself holds great promise as an anti-tumor nanomedicine, and the thorough mechanical insights into NS formation may inspire the design of next-generation multifunctional nanoplatforms.
Collapse
Affiliation(s)
- Bin Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wei Gao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xinyuan Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chunpeng Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Haoyang Du
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yakun Luo
- National Health Commission Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Jiuxin Zhu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, China.
| | - Manjie Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, China.
| |
Collapse
|
2
|
Li SR, Tao SY, Li Q, Hu CY, Sun ZJ. Harnessing nanomaterials for copper-induced cell death. Biomaterials 2025; 313:122805. [PMID: 39250865 DOI: 10.1016/j.biomaterials.2024.122805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/11/2024]
Abstract
Copper (Cu), an essential micronutrient with redox properties, plays a pivotal role in a wide array of pathological and physiological processes across virtually all cell types. Maintaining an optimal copper concentration is critical for cellular survival: insufficient copper levels disrupt respiration and metabolism, while excess copper compromises cell viability, potentially leading to cell death. Similarly, in the context of cancer, copper exhibits a dual role: appropriate amount of copper can promote tumor progression and be an accomplice, yet beyond befitting level, copper can bring about multiple types of cell death, including autophagy, apoptosis, ferroptosis, immunogenic cell death, pyroptosis, and cuproptosis. These forms of cell death are beneficial against cancer progression; however, achieving precise copper regulation within tumors remains a significant challenge in the pursuit of effective cancer therapies. The emergence of nanodrug delivery systems, distinguished by their precise targeting, controlled release, high payload capacity, and the ability to co-deliver multiple agents, has revitalized interest in exploiting copper's precise regulatory capabilities. Nevertheless, there remains a dearth of comprehensive review of copper's bidirectional effects on tumorigenesis and the role of copper-based nanomaterials in modulating tumor progression. This paper aims to address this gap by elucidating the complex role in cancer biology and highlighting its potential as a therapeutic target. Through an exploration of copper's dualistic nature and the application of nanotechnology, this review seeks to offer novel insights and guide future research in advancing cancer treatment.
Collapse
Affiliation(s)
- Su-Ran Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China
| | - Shi-Yue Tao
- Bathune School of Stomatology, Jilin University, Changchun, 130021, Jilin, PR China
| | - Qian Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China
| | - Chuan-Yu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, Hubei, PR China.
| |
Collapse
|
3
|
Zhang W, Song Z, Tian Y, Zhang R, Guo Z, Yang Y, Jiang X, Zhang R. Copper homeostasis and Cuprotosis: Exploring novel therapeutic strategies for connective tissue diseases. Int Immunopharmacol 2025; 145:113698. [PMID: 39642560 DOI: 10.1016/j.intimp.2024.113698] [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: 10/03/2024] [Revised: 11/09/2024] [Accepted: 11/20/2024] [Indexed: 12/09/2024]
Abstract
Copper is an indispensable element for human health, with its balance being critical to prevent the onset of diseases, particularly those affecting connective tissues. Imbalances in copper levels can lead to pathological alterations. Research indicates that copper supplements and chelators hold promise for the treatment of certain conditions, yet the precise mechanisms by which copper imbalances and the cell death mechanism known as cuprotosis contribute to connective tissue diseases remain elusive. This paper delves into the potential role of copper imbalance and cuprotosis in connective tissue diseases and evaluates the underlying cellular mechanisms. The goal is to offer practical insights into targeted therapies for dysregulated copper metabolism, with the aim of devising novel strategies for the treatment of connective tissue diseases.
Collapse
Affiliation(s)
- Wenlan Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhijie Song
- Department of Rheumatology and Immunology, Chifeng Cancer Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Yuanyuan Tian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ruifeng Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhigang Guo
- Department of Rheumatology and Immunology, Chifeng Cancer Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Yanmei Yang
- Department of Rheumatology and Immunology, Chifeng Cancer Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Ruoyi Zhang
- Department of Rheumatology and Immunology, Chifeng Cancer Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
4
|
Dai Y, Zhang Q, Gu R, Chen J, Ye P, Zhu H, Tang M, Nie X. Metal ion formulations for diabetic wound healing: Mechanisms and therapeutic potential. Int J Pharm 2024; 667:124889. [PMID: 39481815 DOI: 10.1016/j.ijpharm.2024.124889] [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: 08/10/2024] [Revised: 10/09/2024] [Accepted: 10/28/2024] [Indexed: 11/03/2024]
Abstract
Metals are vital in human physiology, which not only act as enzyme catalysts in the processes of superoxide dismutase and glucose phosphorylation, but also affect the redox process, osmotic adjustment, metabolism and neural signals. However, metal imbalances can lead to diseases such as diabetes, which is marked by chronic hyperglycemia and affects wound healing. The hyperglycemic milieu of diabetes impairs wound healing, posing significant challenges to patient quality of life. Wound healing encompasses a complex cascade of hemostasis, inflammation, proliferation, and remodeling phases, which are susceptible to disruption in hyperglycemic conditions. In recent decades, metals have emerged as critical facilitators of wound repair by enhancing antimicrobial properties (e.g., iron and silver), providing angiogenic stimulation (copper), promoting antioxidant activity and growth factor synthesis (zinc), and supporting wound closure (calcium and magnesium). Consequently, research has pivoted towards the development of metal ion-based therapeutics, including innovative formulations such as nano-hydrogels, nano-microneedle dressings, and microneedle patches. Prepared by combining macromolecular materials such as chitosan, hyaluronic acid and sodium alginate with metals, aiming at improving the management of diabetic wounds. This review delineates the roles of key metals in human physiology and evaluates the application of metal ions in diabetic wound management strategies.
Collapse
Affiliation(s)
- Yuhe Dai
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Qianbo Zhang
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Rifang Gu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; School Medical Office, Zunyi Medical University, Zunyi 563006, China.
| | - Jitao Chen
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Penghui Ye
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Huan Zhu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Ming Tang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| |
Collapse
|
5
|
Jiang Q, Tong F, Xu Y, Liu C, Xu Q. Cuproptosis: a promising new target for breast cancer therapy. Cancer Cell Int 2024; 24:414. [PMID: 39702350 DOI: 10.1186/s12935-024-03572-2] [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: 06/27/2024] [Accepted: 11/10/2024] [Indexed: 12/21/2024] Open
Abstract
Breast cancer (BC) is the leading cause of cancer-related mortality among women globally, affecting approximately one-quarter of all female cancer patients and accounting for one-sixth of cancer-related deaths in women. Despite significant advancements in diagnostic and therapeutic approaches, breast cancer treatment remains challenging due to issues such as recurrence and metastasis. Recently, a novel form of regulated cell death, termed cuproptosis, has been identified. This process disrupts mitochondrial respiration by targeting the copper-dependent cellular pathways. The role of cuproptosis has been extensively investigated in various therapeutic contexts, including chemotherapy, immunotherapy, radiotherapy, and nanotherapy, with the development of novel drugs significantly improving clinical outcomes. This article aims to further elucidate the connection between cuproptosis and breast cancer, focusing on its therapeutic targets, signaling pathways, and potential biomarkers that could enhance treatment strategies. These insights may offer new opportunities for improved patient care and outcomes in breast cancer therapy.
Collapse
Affiliation(s)
- Qianqian Jiang
- Department of Pharmacy, Traditional Chinese Medicine Hospital of Changshan, Quzhou, 324200, P.R. China
| | - Fei Tong
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, P.R. China
| | - Yun Xu
- Department of Pharmacy, Zhejiang Medical&Health Group Hangzhou Hospital, Hangzhou, Zhejiang, 310022, China
| | - Cheng Liu
- Department of Pharmacy, The Secend People's Hospital Of Jiande, Hangzhou, 311604, P.R. China
| | - Qiaoping Xu
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Cancer Center, Afliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
6
|
Jeena MT, Link J, Zhang J, Harley I, Turunen P, Graf R, Wagner M, Baptista LA, Jonker HRA, Cui L, Lieberwirth I, Landfester K, Rao J, Ng DYW, Weil T. Chaperone-Derived Copper(I)-Binding Peptide Nanofibers Disrupt Copper Homeostasis in Cancer Cells. Angew Chem Int Ed Engl 2024; 63:e202412477. [PMID: 39446574 PMCID: PMC11627128 DOI: 10.1002/anie.202412477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024]
Abstract
Copper (Cu) is a transition metal that plays crucial roles in cellular metabolism. Cu+ homeostasis is upregulated in many cancers and contributes to tumorigenesis. However, therapeutic strategies to target Cu+ homeostasis in cancer cells are rarely explored because small molecule Cu+ chelators have poor binding affinity in comparison to the intracellular Cu+ chaperones, enzymes, or ligands. To address this challenge, we introduce a Cu+ chaperone-inspired supramolecular approach to disrupt Cu+ homeostasis in cancer cells that induces programmed cell death. The Nap-FFMTCGGCR peptide self-assembles into nanofibers inside cancer cells with high binding affinity and selectivity for Cu+ due to the presence of the unique MTCGGC motif, which is conserved in intracellular Cu+ chaperones. Nap-FFMTCGGCR exhibits cytotoxicity towards triple negative breast cancer cells (MDA-MB-231), impairs the activity of Cu+ dependent co-chaperone super oxide dismutase1 (SOD1), and induces oxidative stress. In contrast, Nap-FFMTCGGCR has minimal impact on normal HEK 293T cells. Control peptides show that the self-assembly and Cu+ binding must work in synergy to successfully disrupt Cu+ homeostasis. We show that assembly-enhanced affinity for metal ions opens new therapeutic strategies to address disease-relevant metal ion homeostasis.
Collapse
Affiliation(s)
- M. T. Jeena
- Max-Planck-Institut für PolymerforschungAckermannweg 1055128MainzGermany
| | - Julian Link
- Max-Planck-Institut für PolymerforschungAckermannweg 1055128MainzGermany
| | - Jian Zhang
- Max-Planck-Institut für PolymerforschungAckermannweg 1055128MainzGermany
| | - Iain Harley
- Max-Planck-Institut für PolymerforschungAckermannweg 1055128MainzGermany
| | - Petri Turunen
- Zentrale Einrichtung für MikroskopieInstitut für Molekulare Biologie (IMB)Johannes Gutenberg-UniversitätAckermannweg 455128MainzGermany
| | - Robert Graf
- Max-Planck-Institut für PolymerforschungAckermannweg 1055128MainzGermany
| | - Manfred Wagner
- Max-Planck-Institut für PolymerforschungAckermannweg 1055128MainzGermany
| | | | - Hendrik R. A. Jonker
- Institut für Organische Chemie und Chemische BiologieBiomolekulares Magnetresonanz Zentrum (BMRZ)Goethe Universität Frankfurt60438Frankfurt am MainGermany
| | - Liyang Cui
- Department of RadiologyMolecular Imaging Program at StanfordSchool of MedicineStanford UniversityStanfordCA94305USA
| | - Ingo Lieberwirth
- Max-Planck-Institut für PolymerforschungAckermannweg 1055128MainzGermany
| | | | - Jianghong Rao
- Department of RadiologyMolecular Imaging Program at StanfordSchool of MedicineStanford UniversityStanfordCA94305USA
| | - David Y. W. Ng
- Max-Planck-Institut für PolymerforschungAckermannweg 1055128MainzGermany
| | - Tanja Weil
- Max-Planck-Institut für PolymerforschungAckermannweg 1055128MainzGermany
| |
Collapse
|
7
|
He T, Kang J, Tang X, Wu Y, Hao L. Overexpression of MCL1 attenuates irritable bowel syndrome by regulating cuproptosis: Screening and validation. Biochem Biophys Res Commun 2024; 737:150926. [PMID: 39488085 DOI: 10.1016/j.bbrc.2024.150926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/30/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Irritable bowel syndrome (IBS) is a type of chronic bowel disorder with a poorly understood pathophysiology. Recently, the imbalance of copper has been reported to influence the progression of IBS, suggesting cuproptosis, a new type of copper-induced cell death, may play a role in IBS. This study found 17 cuproptosis-related differentially expressed genes in IBS through bioinformatic analysis. Six hub genes were identified after the protein-protein interaction network analysis, namely myeloid cell leukemia 1 (MCL1), epidermal growth factor receptor 2, cadherin-associated protein beta 1, solute carrier family 25 members 37, solute carrier family 39 members 14, and six transmembrane epithelial antigens of the prostate 3. We selected MCL1 for further verification. Human normal colon epithelial cell line (NCM460) was used to construct models of IBS or cuproptosis in vitro by lipopolysaccharide (LPS) or LPS combined with copper (II) chloride (CuCl2). We observed that overexpression of MCL1 promoted cell viability and proliferation ability, and inhibited the secretion of inflammatory factors and expression of Bax and caspase-3 of NCM460 cells treated with LPS or LPS combined with CuCl2. In addition, up-regulated MCL1 significantly suppressed the protein levels of ferredoxin 1 and lipoyl synthase, two key regulators of cuproptosis. In conclusion, our study demonstrates that cuproptosis is involved in IBS and identifies a cuproptosis-related gene, MCL1, that helps alleviate IBS by promoting cell growth, reducing inflammation, and suppressing cuproptosis, making it a promising therapeutic target in IBS.
Collapse
Affiliation(s)
- Taohong He
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610072, China
| | - Jian Kang
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610072, China; School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, No.37-39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610075, China
| | - Xiao Tang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No. 23, Meishuguan Back Street, Dongcheng District, Beijing, 100010, China
| | - Yuqi Wu
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610072, China; School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, No.37-39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610075, China
| | - Liangliang Hao
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39, Shi-er-qiao Road, Jinniu District, Chengdu City, Sichuan Province, 610072, China.
| |
Collapse
|
8
|
Pushie MJ, Sylvain NJ, Hou H, Pendleton N, Wang R, Zimmermann L, Pally M, Cayabyab FS, Peeling L, Kelly ME. X-ray fluorescence mapping of brain tissue reveals the profound extent of trace element dysregulation in stroke pathophysiology. Metallomics 2024; 16:mfae054. [PMID: 39547935 PMCID: PMC11631071 DOI: 10.1093/mtomcs/mfae054] [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: 06/18/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024]
Abstract
The brain is a privileged organ with regard to its trace element composition and maintains a robust barrier system to sequester this specialized environment from the rest of the body and the vascular system. Stroke is caused by loss of adequate blood flow to a region of the brain. Without adequate blood flow ischaemic changes begin almost immediately, triggering an ischaemic cascade, characterized by ion dysregulation, loss of function, oxidative damage, cellular degradation, and breakdown of the barrier that helps maintain this environment. Ion dysregulation is a hallmark of stroke pathophysiology and we observe that most elements in the brain are dysregulated after stroke. X-ray fluorescence-based detection of physiological changes in the neurometallome after stroke reveals profound ion dysregulation within the lesion and surrounding tissue. Not only are most elements significantly dysregulated after stroke, but the level of dysregulation cannot be predicted from a cell-level description of dysregulation. X-ray fluorescence imaging reveals that the stroke lesion retains <25% of essential K+ after stroke, but this element is not concomitantly elevated elsewhere in the organ. Moreover, elements like Na+, Ca2+, and Cl- are vastly elevated above levels available in normal brain tissue (>400%, >200%, and >150%, respectively). We hypothesize that weakening of the blood-brain barrier after stroke allows elements to freely diffuse down their concentration gradient so that the stroke lesion is in equilibrium with blood (and the compartments containing brain interstitial fluid and cerebrospinal fluid). The change observed for the neurometallome likely has consequences for the potential to rescue infarcted tissue, but also presents specific targets for treatment.
Collapse
Affiliation(s)
- M Jake Pushie
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Nicole J Sylvain
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Huishu Hou
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Nicole Pendleton
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Richard Wang
- College of Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Liam Zimmermann
- College of Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Maxwell Pally
- College of Arts & Science, Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Francisco S Cayabyab
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Lissa Peeling
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Michael E Kelly
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
9
|
Cui Y, Du X, Li Y, Wang D, Lv Z, Yuan H, Chen Y, Liu J, Sun Y, Wang W. Imbalanced and Unchecked: The Role of Metal Dyshomeostasis in Driving COPD Progression. COPD 2024; 21:2322605. [PMID: 38591165 DOI: 10.1080/15412555.2024.2322605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/19/2024] [Indexed: 04/10/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic respiratory condition characterized by persistent inflammation and oxidative stress, which ultimately leads to progressive restriction of airflow. Extensive research findings have cogently suggested that the dysregulation of essential transition metal ions, notably iron, copper, and zinc, stands as a critical nexus in the perpetuation of inflammatory processes and oxidative damage within the lungs of COPD patients. Unraveling the intricate interplay between metal homeostasis, oxidative stress, and inflammatory signaling is of paramount importance in unraveling the intricacies of COPD pathogenesis. This comprehensive review aims to examine the current literature on the sources, regulation, and mechanisms by which metal dyshomeostasis contributes to COPD progression. We specifically focus on iron, copper, and zinc, given their well-characterized roles in orchestrating cytokine production, immune cell function, antioxidant depletion, and matrix remodeling. Despite the limited number of clinical trials investigating metal modulation in COPD, the advent of emerging methodologies tailored to monitor metal fluxes and gauge responses to chelation and supplementation hold great promise in unlocking the potential of metal-based interventions. We conclude that targeted restoration of metal homeostasis represents a promising frontier for ameliorating pathological processes driving COPD progression.
Collapse
Affiliation(s)
- Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xinqian Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yunqi Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Dan Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Huihui Yuan
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Jie Liu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ying Sun
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
10
|
Guo K, Wang T, Yin J, Yang S, Cui H, Cao Z, Zhao Q, Xie G, Lu J, Gu G, Wu W. Identification of Cuproptosis-Related Patterns Predict Prognosis and Immunotherapy Response in Hepatocellular Carcinoma. J Cell Mol Med 2024; 28:e70224. [PMID: 39663596 PMCID: PMC11634814 DOI: 10.1111/jcmm.70224] [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: 09/18/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024] Open
Abstract
A novel copper-dependent mode of death, cuproptosis, has been newly identified. This study developed a cuproptosis score (CS) based on the cuproptosis model to analyse the association of CS with prognosis, immune cell infiltration, drug sensitivity and immunotherapy response in hepatocellular carcinoma (HCC) patients. A typing model of cuproptosis was constructed based on the expression of 19 cuproptosis-related genes (CRGs). A total of 485 samples were divided into high scoring group (HSG) and low scoring group (LSG) according to CS, and the drug sensitivity and responsiveness to immunotherapy were evaluated by combining the immunophenotype score (IPS), oncoPredict, the tumour immune dysfunction and rejection (TIDE). The use of weighted gene coexpression network analysis (WGCNA) identified key prognostic genes for cuproptosis. Western blotting was used to detect the expression level of the key gene. The CRG key gene glutaminase (GLS) is highly expressed in HCC, and patients with high expression of GLS have a poorer prognosis. Furthermore, cell function experiments, such as proliferation, migration and invasion assays, confirmed that GLS knockdown significantly changed the incidence and progression of HCC. This study suggests that new biological markers associated with cuproptosis can be used in the clinical diagnosis of HCC patients to predict prognosis and therapeutic targets.
Collapse
Affiliation(s)
- Kai Guo
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
| | - Tianbing Wang
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
| | - Jimin Yin
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
- Anhui No.2 Provincial People's HospitalHefeiChina
- The Fifth Clinical Medical College of Anhui Medical UniversityHefeiChina
| | - Shoushan Yang
- Department of General SurgeryThe Fourth People's Hospital of Lu'anChina
| | - Haodong Cui
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
- Anhui No.2 Provincial People's HospitalHefeiChina
- The Fifth Clinical Medical College of Anhui Medical UniversityHefeiChina
| | - Zichuan Cao
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
| | - Qiang Zhao
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
| | - Gongbo Xie
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
| | - Jian Lu
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
| | - Guosheng Gu
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
- Anhui No.2 Provincial People's HospitalHefeiChina
| | - Wenyong Wu
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
- Anhui No.2 Provincial People's HospitalHefeiChina
- The Fifth Clinical Medical College of Anhui Medical UniversityHefeiChina
| |
Collapse
|
11
|
Zheng YL, Ji Y, Li Y, Yan S, Ren XR, Tang W, Dai F, Zhou B. Identification of tanshinone I as a natural Cu(II) ionophore. Free Radic Biol Med 2024; 227:27-41. [PMID: 39613045 DOI: 10.1016/j.freeradbiomed.2024.11.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/11/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
The development of Cu(II) ionophores for targeted disruption of aberrant redox homeostasis in cancer cells has been considered an appealing strategy in the field of anticancer research. This study presents the first identification of tanshinone I (Ts1), a natural o-quinone, as a Cu(II) ionophore. Structure-activity relationship studies on tanshinones and mechanistic investigations reveal that the presence of Cu(II) effectively promotes the tautomerization of Ts1 from its diketo to keto-enol forms, thereby facilitating its sequential proton-loss Cu(II) chelation, and enabling it to function as a Cu(II) ionophore due to its structural features including the presence of an o-quinone moiety, a benzyl hydrogen, and a large conjugated system. The unique property allows Ts1 to preferentially induce copper accumulation in human hepatoma HepG2 cells over human umbilical vein endothelial cells, by releasing copper driven by reduced glutathione (GSH). This copper accumulation leads to a reduction in the GSH-to-oxidized glutathione ratio and the generation of reactive oxygen species, ultimately triggering apoptosis of HepG2 cells. The findings not only provide support for o-quinones as innovative types of anticancer Cu(II) ionophores, but also shed light on the previously unrecognized role of Ts1 as a potent Cu(II) ionophore for eradicating cancer cells by selectively disrupting their redox regulation programs, resembling a "Trojan horse".
Collapse
Affiliation(s)
- Ya-Long Zheng
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., 730000, Lanzhou, Gansu, China; Medicine College of Pingdingshan University, 467000, Pingdingshan, Henan, China
| | - Yuan Ji
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., 730000, Lanzhou, Gansu, China
| | - Yan Li
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., 730000, Lanzhou, Gansu, China
| | - Shuai Yan
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., 730000, Lanzhou, Gansu, China
| | - Xiao-Rong Ren
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., 730000, Lanzhou, Gansu, China
| | - Wei Tang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., 730000, Lanzhou, Gansu, China
| | - Fang Dai
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., 730000, Lanzhou, Gansu, China.
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., 730000, Lanzhou, Gansu, China.
| |
Collapse
|
12
|
Guo D, Huang Z, Wang Q, Chen W, Huang Y, Sun X, Chen J, Feng S. IAPP blocks anti-breast cancer function of CD8 +T cells via targeting cuproptosis. Front Immunol 2024; 15:1481129. [PMID: 39654888 PMCID: PMC11625781 DOI: 10.3389/fimmu.2024.1481129] [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: 08/15/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Background Breast cancer (BRCA) is the most prevalent type of cancer worldwide. As a highly heterogeneous cancer, it has a high recurrence rate. Since its biological behavior can be regulated by immunity and cuprotosis, so exploring potential therapeutic target to mediate immunity and cuprotosis is of great significance for BRCA therapy. Methods The immune-related genes and immune-cuprotosis-related deferentially expressed genes (ICR-DEGs) were identified by mining the TCGA database. Prognostic analysis, differential expression analysis, univariate and lasso regression analyses were used to determine their independent prognostic values. To evaluate the relationship between ICR-DEGs and immune scores, we constructed a prognostic risk model to evaluate immune checkpoints, and then the role of tumor immune microenvironment in BRCA was explored. Furthermore, anti-BRCA function and mechanism of islet amyloid poly-peptide (IAPP) mediated CD8+T cells were verified by means of flow cytometry, ELISA, and subcutaneous transplantation tumor model. Results All results suggested that immune-cuprotosis-related genes were a potential predictor of BRCA's response to immune checkpoint inhibitors and immunotherapy biomarkers. Thereby downregulation of IAPP reduced cuprotosis of CD8+T or Her2-CAR-T cells to promote the anti-BRCA function both in vitro and in vivo. Conclusion Our research had clarified the function and mechanism of IAPP in CD8+T cells, providing new ideas for improving the diagnosis and treatment of BRCA.
Collapse
Affiliation(s)
- Dandan Guo
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou, China
| | - Zhijian Huang
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Qianqian Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou, China
| | - Wei Chen
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou, China
| | - Yu Huang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou, China
| | - Xinhao Sun
- Department of Thyroid and Breast Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, China
| | - Jian Chen
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Shuying Feng
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou, China
| |
Collapse
|
13
|
Zhou M, Tang J, Huang G, Hong L. Prognostic Significance and Immune Landscape of a Cuproptosis-Related LncRNA Signature in Ovarian Cancer. Biomedicines 2024; 12:2640. [PMID: 39595204 PMCID: PMC11592286 DOI: 10.3390/biomedicines12112640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Cuproptosis is a copper-induced mitochondrial cell death, and regulating cuproptosis is becoming a rising cancer treatment modality. Here, we attempted to establish a cuproptosis-associated lncRNAs (CRLs) signature (CRlncSig) to predict the survival, immune landscape, and treatment response in ovarian cancer (OC) patients. Methods: A series of statistical analyses were used to identify the key CRLs that are closely related to the prognosis, and a prognostic CRlncSig was constructed. The predictive accuracy of the CRlncSig was further validated in an independent Gene Expression Omnibus (GEO) set. Then, we compared the immune cell infiltration, immune checkpoints, tumor microenvironment (TME), tumor mutational burden (TMB), drug sensitivity, and efficacy of immunotherapy between the two subgroups. We further built a nomogram integrating the CRlncSig and different clinical traits to enhance the clinical application of the CRlncSig. Results: Nine hub CRLs, namely RGMB-AS1, TYMSOS, DANCR, LINC00702, LINC00240, LINC00996, DNM1P35, LINC00892, and TMEM254-AS1, were correlated with the overall survival (OS) of OC and a prognostic CRlncSig was established. The CRlncSig classified OC patients into two risk groups with strikingly different survival probabilities. The time-dependent ROC (tdROC) curves demonstrated good predictive ability in both the training cohort and an independent validation cohort. Multivariate analysis confirmed the independent predictive performance of the CRlncSig. We constructed a nomogram based on the CRlncSig, which can predict the prognosis of OC patients. The high-risk score was characterized by decreased immune cell infiltration and activation of stroma, while activation of immunity was observed in the low-risk subgroup. Moreover, patients in low-risk subgroups had more Immunophenoscore (IPS) and fewer immune escapes compared to high-risk subgroups. Finally, an immunotherapeutic cohort confirmed the value of the CRlncSig in predicting immunotherapy outcomes. Conclusions: The developed CRlncSig may be promising for the clinical prediction of OC patient outcomes and immunotherapeutic responses.
Collapse
Affiliation(s)
| | | | | | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (M.Z.)
| |
Collapse
|
14
|
Ju S, Singh MK, Han S, Ranbhise J, Ha J, Choe W, Yoon KS, Yeo SG, Kim SS, Kang I. Oxidative Stress and Cancer Therapy: Controlling Cancer Cells Using Reactive Oxygen Species. Int J Mol Sci 2024; 25:12387. [PMID: 39596452 PMCID: PMC11595237 DOI: 10.3390/ijms252212387] [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: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer is a multifaceted disease influenced by various mechanisms, including the generation of reactive oxygen species (ROS), which have a paradoxical role in both promoting cancer progression and serving as targets for therapeutic interventions. At low concentrations, ROS serve as signaling agents that enhance cancer cell proliferation, migration, and resistance to drugs. However, at elevated levels, ROS induce oxidative stress, causing damage to biomolecules and leading to cell death. Cancer cells have developed mechanisms to manage ROS levels, including activating pathways such as NRF2, NF-κB, and PI3K/Akt. This review explores the relationship between ROS and cancer, focusing on cell death mechanisms like apoptosis, ferroptosis, and autophagy, highlighting the potential therapeutic strategies that exploit ROS to target cancer cells.
Collapse
Affiliation(s)
- Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jyotsna Ranbhise
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Geun Yeo
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02453, Republic of Korea;
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
15
|
Zhang L, Xie A, Ma J, Liu H, Zeng C. Unveiling Cuproptosis: Mechanistic insights, roles, and leading advances in oncology. Biochim Biophys Acta Rev Cancer 2024; 1879:189180. [PMID: 39276875 DOI: 10.1016/j.bbcan.2024.189180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024]
Abstract
Copper, a vital micronutrient, performs essential functions in numerous biological settings. Its disrupted metabolism is implicated in both the initiation of tumors and therapeutic interventions for cancer, underscoring the critical necessity of preserving copper homeostasis. Cuproptosis, a regulated cell death (RCD) modulated by copper, is activated in response to elevated copper concentrations, prompting an investigation into its implication in oncogenesis. Within this review, an exploration is conducted into copper dynamics and homeostasis maintenance within cells. Furthermore, it delves into the mechanisms underlying cuproptosis and its interplay with signaling pathways implicated in cancer. The potential synergy between cuproptosis and ferroptosis and its impact on tumor immunomodulation is discussed. Additionally, promising avenues for addressing cuproptosis in cancer involve assessing the utility of copper chelators and ionophores. By addressing pressing questions surrounding cuproptosis and outlining its pivotal role in cancer pathogenesis and treatment, this review propounds targeting cuproptosis as a promising frontier in antitumor therapy, potentially revolutionizing cancer treatment strategies.
Collapse
Affiliation(s)
- Limei Zhang
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Aihui Xie
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Jingxian Ma
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Huilin Liu
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen 518110, China.
| |
Collapse
|
16
|
Villatoro-Santos CR, Ramirez-Zea M, Villamor E. Plasma Copper and Metabolic Syndrome in Mesoamerican Children and Their Parents. Biol Trace Elem Res 2024; 202:4903-4908. [PMID: 38240932 DOI: 10.1007/s12011-024-04069-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/15/2024] [Indexed: 10/01/2024]
Abstract
The role of copper in the etiology of metabolic syndrome (MetS) is uncertain. We evaluated associations of plasma copper concentrations with MetS and its components in a cross-sectional study of 198 children ages 7-12 years and 378 adult parents from eight Mesoamerican countries. In children, the outcome was a metabolic risk score based on waist circumference, insulin resistance, mean arterial pressure (MAP), and blood lipids. In adults, we defined MetS per Adult Treatment Panel III criteria. Plasma copper was not significantly related to MetS in children or adults; however, children with copper above the median had a MAP score 0.04 (95% CI, 0.002, 0.08; P = 0.04) adjusted units higher and a HDL-cholesterol score 0.07 (95% CI, - 0.13, - 0.003; P = 0.04) adjusted units lower than those with lower copper concentrations. In adults, copper was positively related to abdominal obesity. Longitudinal studies to confirm the deleterious role of copper on MetS components are warranted.
Collapse
Affiliation(s)
- Claudia R Villatoro-Santos
- Department of Epidemiology, University of Michigan School of Public Health, 1415 Washington Heights, SPH II, Room M5507, Ann Arbor, MI, 48109, USA
| | - Manuel Ramirez-Zea
- Institute of Nutrition of Central America and Panama, INCAP Research Center for the Prevention of Chronic Diseases, Guatemala City, Guatemala
| | - Eduardo Villamor
- Department of Epidemiology, University of Michigan School of Public Health, 1415 Washington Heights, SPH II, Room M5507, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
17
|
Wang YM, Feng LS, Xu A, Ma XH, Zhang MT, Zhang J. Copper ions: The invisible killer of cardiovascular disease (Review). Mol Med Rep 2024; 30:210. [PMID: 39301641 PMCID: PMC11425066 DOI: 10.3892/mmr.2024.13334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
Copper, a vital trace element, is indispensable for the maintenance of physiological functioning, particularly in the cardiac system. Unlike other forms of cell death such as iron death and apoptosis, copper‑induced cell death has gained increasing recognition as a significant process influencing the development of cardiovascular diseases. The present review highlights the significance of maintaining copper homeostasis in addressing cardiovascular diseases. This review delves into the crucial roles of copper in physiology, including the metabolic pathways and its absorption, transport and excretion. It provides detailed insights into the mechanisms underlying cardiovascular diseases resulting from both excess and deficient copper levels. Additionally, it summarizes strategies for treating copper imbalances through approaches such as copper chelators and ion carriers while discussing their limitations and future prospects.
Collapse
Affiliation(s)
- Yi-Ming Wang
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Lan-Shuan Feng
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Ao Xu
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Xiao-Han Ma
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Miao-Tiao Zhang
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Jie Zhang
- Cardiovascular Department, Xi'an Fifth Hospital, Xi'an, Shaanxi 710000, P.R. China
| |
Collapse
|
18
|
Sheng H, Gu J, Huang Y, Kołat D, Shi G, Yan L, Ye D. Cuproptosis-related signature predicts prognosis and indicates tumor immune infiltration in bladder cancer. Transl Androl Urol 2024; 13:2280-2293. [PMID: 39507864 PMCID: PMC11535731 DOI: 10.21037/tau-24-456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/29/2024] [Indexed: 11/08/2024] Open
Abstract
Background Cuproptosis is a newly identified form of cell death that is dependent on copper (Cu) ions, termed Cu-dependent cytotoxicity. This process is distinct from other forms of cell death such as apoptosis, necrosis, and ferroptosis. The accumulation of copper is known to play a significant role in various biological processes, including angiogenesis (the formation of new blood vessels) and metastasis (the spread of cancer cells to different parts of the body). These processes are crucial for tumor growth and progression, indicating that copper and the cuproptosis-related genes (CPRGs) might be indispensable in the context of cancer development and progression. Given this background, we aimed to explore the relationship between CPRGs and both prognostic predictions and tumor microenvironment (TME) infiltration in bladder cancer (BLCA). Methods For this study, we utilized data from The Cancer Genome Atlas (TCGA) to identify CPRGs and subsequently divided BLCA patients into three distinct molecular clusters based on these genes. To assess the proportions of various immune cell types within the TME, we employed single-sample gene set enrichment analysis (ssGSEA) and the Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) method. These computational techniques allowed us to quantify the infiltration of different immune cells, providing insights into the immune landscape of the tumors. Furthermore, we developed a risk score model using CPRGs to predict the survival prospects of BLCA patients. Results Our analysis identified three molecular clusters of BLCA patients, each exhibiting unique clinical features and patterns of TME infiltration. Among these clusters, cluster 1 was associated with a poor prognosis. Interestingly, this cluster also showed significant infiltration of activated CD4+ (ssGSEA P<0.001) and CD8+ T (ssGSEA P<0.05) cells, which are crucial components of the immune response against tumors. This finding suggests a complex interaction between the immune system and the tumor, where a high presence of T cells does not necessarily correlate with better outcomes. Additionally, our risk score model revealed that the high-risk group, characterized by a specific expression pattern of CPRGs, also had enhanced infiltration of CD4+ and CD8+ T cells. This indicates that the cuproptosis-based risk model has a robust ability to predict patient prognosis and can guide immunotherapy decisions. Conclusions Our study sheds light on the biological functions of CPRGs within the TME of BLCA and their correlations with clinical parameters and patient prognosis. The identification of distinct molecular clusters with varying prognoses and immune cell infiltrations highlights the heterogeneity of BLCA and underscores the potential of CPRGs as biomarkers for prognosis and therapeutic targets. These findings offer new perspectives for the development of immunotherapeutic strategies in the treatment of BLCA patients, potentially leading to more personalized and effective cancer therapies.
Collapse
Affiliation(s)
- Haoyue Sheng
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiani Gu
- Department of Nursing, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yongqiang Huang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Damian Kołat
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz, Poland
- Department of Functional Genomics, Medical University of Lodz, Lodz, Poland
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lihua Yan
- Department of Nursing, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Ghai S, Shrestha R, Hegazi A, Boualoy V, Liu SH, Su KH. The Role of Heat Shock Factor 1 in Preserving Proteomic Integrity During Copper-Induced Cellular Toxicity. Int J Mol Sci 2024; 25:11657. [PMID: 39519208 PMCID: PMC11546224 DOI: 10.3390/ijms252111657] [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: 10/04/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Copper is crucial for many physiological processes across mammalian cells, including energy metabolism, neurotransmitter synthesis, and antioxidant defense mechanisms. However, excessive copper levels can lead to cellular toxicity and "cuproptosis", a form of programmed cell death characterized by the accumulation of copper within mitochondria. Tumor cells are less sensitive to this toxicity than normal cells, the mechanism for which remains unclear. We address this important issue by exploring the role of heat shock factor 1 (HSF1), a transcription factor that is highly expressed across several types of cancer and has a crucial role in tumor survival, in protecting against copper-mediated cytotoxicity. Using pancreatic ductal adenocarcinoma cells, we show that excessive copper triggers a proteotoxic stress response (PSR), activating HSF1 and that overexpressing HSF1 diminishes intracellular copper accumulation and prevents excessive copper-induced cell death and amyloid fibrils formation, highlighting HSF1's role in preserving proteasomal integrity. Copper treatment decreases the lipoylation of dihydrolipoamide S-acetyltransferase (DLAT), an enzyme necessary for cuproptosis, induces DLAT oligomerization, and induces insoluble DLAT formation, which is suppressed by overexpressing HSF1, in addition to enhancing the interaction between HSF1 and DLAT. Our findings uncover how HSF1 protects against copper-induced damage in cancer cells and thus represents a novel therapeutic target for enhancing copper-mediated cancer cell death.
Collapse
Affiliation(s)
| | | | | | | | | | - Kuo-Hui Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (S.G.); (R.S.); (A.H.); (V.B.); (S.-H.L.)
| |
Collapse
|
20
|
Wróblewska J, Nuszkiewicz J, Wróblewski M, Wróblewska W, Woźniak A. Selected Trace Elements and Their Impact on Redox Homeostasis in Eye Health. Biomolecules 2024; 14:1356. [PMID: 39595533 PMCID: PMC11591929 DOI: 10.3390/biom14111356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Oxidative stress plays a crucial role in the pathogenesis of various ocular degenerative diseases, leading to structural and functional changes in eye tissues. This imbalance between reactive oxygen species (ROS) and antioxidants significantly contributes to conditions such as age-related macular degeneration, diabetic retinopathy, cataracts, and glaucoma. Both enzymatic and nonenzymatic antioxidants are vital for maintaining ocular health by neutralizing ROS and restoring cellular redox balance. Essential trace elements, including iron, zinc, copper, and selenium, are fundamental for the proper functioning of these antioxidant systems. Iron is indispensable for enzymatic activity and cellular energy production, zinc supports numerous proteins involved in visual functions and antioxidant defense, copper is essential for various enzymatic reactions preventing oxidative stress, and selenium is critical for the activity of antioxidant enzymes such as glutathione peroxidase (GPX) and thioredoxin reductase (TrxR). This review summarizes current research on the complex interactions between oxidative stress and trace elements in ocular diseases, highlighting the therapeutic potential of antioxidant supplementation to mitigate oxidative damage and improve eye health. By integrating insights from studies on oxidative stress, trace elements, and eye physiology, this article underscores new diagnostic and therapeutic strategies that could lead to more effective prevention and treatment of ocular diseases, aiming to enhance clinical outcomes and guide future research in optimizing therapeutic strategies for eye health.
Collapse
Affiliation(s)
- Joanna Wróblewska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland; (J.W.); (M.W.)
| | - Jarosław Nuszkiewicz
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland; (J.W.); (M.W.)
| | - Marcin Wróblewski
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland; (J.W.); (M.W.)
| | - Weronika Wróblewska
- Student Research Club of Medical Biology and Biochemistry, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland;
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland; (J.W.); (M.W.)
| |
Collapse
|
21
|
Noh D, Lee H, Lee S, Sun IC, Yoon HY. Copper-Based Nanomedicines for Cuproptosis-Mediated Effective Cancer Treatment. Biomater Res 2024; 28:0094. [PMID: 39430913 PMCID: PMC11486892 DOI: 10.34133/bmr.0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
The recent discovery of cuproptosis, a novel copper-ion-induced cell death pathway, has suggested the novel therapeutic potential for treating heterogeneous and drug-resistant cancers. Currently, copper ionophore-based therapeutics have been designed to treat cancers, utilizing copper ions as a strategic tool to impede tumor proliferation and promote cellular demise. However, limitations of copper ionophore-based therapies include nontargeted delivery of copper ions, low tumor accumulation, and short half-life. Strategies to enhance specificity involve targeting intracellular cuproptosis mechanisms using nanotechnology-based drugs. Additionally, the importance of exploring combination therapies cannot be overstated, as they are a key strategy in improving the efficacy of cancer treatments. Recent studies have reported the anticancer effects of nanomedicines that can induce cuproptosis of cancer both in vitro and in vivo. These cuproptosis-targeted nanomedicines could improve delivery efficiency with the pharmacokinetic properties of copper ion, resulting in increasing cuproptosis-based anticancer effects. This review will summarize the intricate nexus between copper ion and carcinogenesis, examining the pivotal roles of copper homeostasis and its dysregulation in cancer progression and fatality. Furthermore, we will introduce the latest advances in cuproptosis-targeted nanomedicines for cancer treatment. Finally, the challenges in cuproptosis-based nanomedicines will be discussed for future development directions.
Collapse
Affiliation(s)
- Dahye Noh
- Medicinal Materials Research Center, Biomedical Research Institute,
Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School,
University of Science and Technology (UST), Hwarang-ro14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hokyung Lee
- Medicinal Materials Research Center, Biomedical Research Institute,
Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- Department of Fundamental Pharmaceutical Sciences, College of Pharmacy,
Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sangmin Lee
- Department of Fundamental Pharmaceutical Sciences, College of Pharmacy,
Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - In-Cheol Sun
- Medicinal Materials Research Center, Biomedical Research Institute,
Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hong Yeol Yoon
- Medicinal Materials Research Center, Biomedical Research Institute,
Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School,
University of Science and Technology (UST), Hwarang-ro14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
22
|
Chang H, Zhang W, Xu L, Li Z, Lin C, Shen Y, Zhang G, Mao L, Ma C, Liu N, Lu H. Copper aggravated synaptic damage after traumatic brain injury by downregulating BNIP3-mediated mitophagy. Autophagy 2024:1-17. [PMID: 39415457 DOI: 10.1080/15548627.2024.2409613] [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: 07/12/2023] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/18/2024] Open
Abstract
Synaptic damage is a crucial pathological process in traumatic brain injury. However, the mechanisms driving this process remain poorly understood. In this report, we demonstrate that the accumulation of damaged mitochondria, resulting from impaired mitphagy, plays a significant role in causing synaptic damage. Moreover, copper induced downregulation of BNIP3 is a key player in regulating mitophagy. DMSA alleviates synaptic damage and mitochondrial dysfunction by promoting urinary excretion of copper. Mechanistically, we find that copper downregulate BNIP3 by increasing the nuclear translocation of NFKB, which is triggered by TRIM25-mediated ubiquitination-dependent degradation of NFKBIA. Our study underscores the importance of copper accumulation in the regulation of BNIP3-mediated mitophagy and suggests that therapeutic targeting of the copper-TRIM25-NFKB-BNIP3 axis holds promise to attenuate synaptic damage after traumatic brain injury.
Collapse
Affiliation(s)
- Hanxiao Chang
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Weiwei Zhang
- Department of Ophthalmology, Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lei Xu
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Zheng Li
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Chao Lin
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Yuqi Shen
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Guangjian Zhang
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Lei Mao
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Chencheng Ma
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Ning Liu
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Hua Lu
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
23
|
Murata D, Roy S, Lutsenko S, Iijima M, Sesaki H. Slc25a3-dependent copper transport controls flickering-induced Opa1 processing for mitochondrial safeguard. Dev Cell 2024; 59:2578-2592.e7. [PMID: 38986607 PMCID: PMC11461135 DOI: 10.1016/j.devcel.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/18/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
Following the Goldilocks principle, mitochondria size must be "just right." Mitochondria balance division and fusion to avoid becoming too big or too small. Defects in this balance produce dysfunctional mitochondria in human diseases. Mitochondrial safeguard (MitoSafe) is a defense mechanism that protects mitochondria against extreme enlarging by suppressing fusion in mammalian cells. In MitoSafe, hyperfused mitochondria elicit flickering-short pulses of mitochondrial depolarization. Flickering activates an inner membrane protease, Oma1, which in turn proteolytically inactivates a mitochondrial fusion protein, Opa1. The mechanisms underlying flickering are unknown. Using a live-imaging screen, we identified Slc25a3 (a mitochondrial carrier transporting phosphate and copper) as necessary for flickering and Opa1 cleavage. Remarkably, copper, but not phosphate, is critical for flickering. Furthermore, we found that two copper-containing mitochondrial enzymes, superoxide dismutase 1 and cytochrome c oxidase, regulate flickering. Our data identify an unforeseen mechanism linking copper, redox homeostasis, and membrane flickering in mitochondrial defense against deleterious fusion.
Collapse
Affiliation(s)
- Daisuke Murata
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shubhrajit Roy
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Svetlana Lutsenko
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
24
|
Huang Q, Huang XY, Xue YT, Wu XH, Wu YP, Ke ZB, Kang Z, Xu YC, Chen DN, Wei Y, Xue XY, Huang ZY, Xu N. Molecular Subtypes Defined by Cuproptosis-Associated Genes, Prognostic Model Development, and Tumor Immune Microenvironment Characterization in Adrenocortical Carcinoma. J Inflamm Res 2024; 17:7017-7036. [PMID: 39377045 PMCID: PMC11457769 DOI: 10.2147/jir.s461489] [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: 04/22/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024] Open
Abstract
Introduction This study aims to explore the role of cuproptosis-related genes in ACC, utilizing data from TCGA and GEO repositories, and to develop a predictive model for patient stratification. Methods A cohort of 123 ACC patients with survival data was analyzed. RNA-seq data of 17 CRGs were examined, and univariate Cox regression identified prognostic CRGs. A cuproptosis-related network was constructed to show interactions between CRGs. Consensus clustering classified ACC into three subtypes, with transcriptional and survival differences assessed by PCA and survival analysis. Gene set variation analysis (GSVA) and ssGSEA evaluated functional and immune infiltration characteristics across subtypes. Differentially expressed genes (DEGs) were identified, and gene clusters were established. A risk score (CRG_score) was generated using LASSO and multivariate Cox regression, validated across datasets. Tumor microenvironment, stem cell index, mutation status, drug sensitivity, and hormone synthesis were examined in relation to the CRG_score. Protein expression of key genes was validated, and functional studies on ASF1B and NDRG4 were performed. Results Three ACC subtypes were identified with distinct survival outcomes. Subtype B showed the worst prognosis, while subtype C had the best. We identified 214 DEGs linked to cell proliferation and classified patients into three gene clusters, confirming their prognostic value. The CRG_score predicted patient outcomes, with high-risk patients demonstrating worse survival and possible resistance to immunotherapy. Drug sensitivity analysis suggested higher responsiveness to doxorubicin and etoposide in high-risk patients. Conclusion This study suggests the potential prognostic value of CRGs in ACC. The CRG_score model provides a robust tool for risk stratification, with implications for treatment strategies.
Collapse
Affiliation(s)
- Qi Huang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
- Department of Urology, Quanzhou First Hospital, Fujian Medical University, Quanzhou, 362000, People’s Republic of China
| | - Xu-Yun Huang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
| | - Yu-Ting Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
| | - Xiao-Hui Wu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
| | - Yu-Peng Wu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
| | - Zhi-Bin Ke
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
| | - Zhen Kang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
| | - Yi-Cheng Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
| | - Dong-Ning Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
| | - Yong Wei
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
| | - Zhi-Yang Huang
- Department of Urology, Quanzhou First Hospital, Fujian Medical University, Quanzhou, 362000, People’s Republic of China
| | - Ning Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| |
Collapse
|
25
|
Min JH, Sarlus H, Harris RA. MAD-microbial (origin of) Alzheimer's disease hypothesis: from infection and the antimicrobial response to disruption of key copper-based systems. Front Neurosci 2024; 18:1467333. [PMID: 39416952 PMCID: PMC11480022 DOI: 10.3389/fnins.2024.1467333] [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: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Microbes have been suspected to cause Alzheimer's disease since at least 1908, but this has generally remained unpopular in comparison to the amyloid hypothesis and the dominance of Aβ and Tau. However, evidence has been accumulating to suggest that these earlier theories are but a manifestation of a common cause that can trigger and interact with all the major molecular players recognized in AD. Aβ, Tau and ApoE, in particular appear to be molecules with normal homeostatic functions but also with alternative antimicrobial functions. Their alternative functions confer the non-immune specialized neuron with some innate intracellular defenses that appear to be re-appropriated from their normal functions in times of need. Indeed, signs of infection of the neurons by biofilm-forming microbial colonies, in synergy with herpes viruses, are evident from the clinical and preclinical studies we discuss. Furthermore, we attempt to provide a mechanistic understanding of the AD landscape by discussing the antimicrobial effect of Aβ, Tau and ApoE and Lactoferrin in AD, and a possible mechanistic link with deficiency of vital copper-based systems. In particular, we focus on mitochondrial oxidative respiration via complex 4 and ceruloplasmin for iron homeostasis, and how this is similar and possibly central to neurodegenerative diseases in general. In the case of AD, we provide evidence for the microbial Alzheimer's disease (MAD) theory, namely that AD could in fact be caused by a long-term microbial exposure or even long-term infection of the neurons themselves that results in a costly prolonged antimicrobial response that disrupts copper-based systems that govern neurotransmission, iron homeostasis and respiration. Finally, we discuss potential treatment modalities based on this holistic understanding of AD that incorporates the many separate and seemingly conflicting theories. If the MAD theory is correct, then the reduction of microbial exposure through use of broad antimicrobial and anti-inflammatory treatments could potentially alleviate AD although this requires further clinical investigation.
Collapse
Affiliation(s)
- Jin-Hong Min
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital at Solna, Stockholm, Sweden
| | | | | |
Collapse
|
26
|
Focaccio A, Rossi L, De Luca A. A spotlight on the role of copper in the epithelial to mesenchymal transition. Life Sci 2024; 354:122972. [PMID: 39142503 DOI: 10.1016/j.lfs.2024.122972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/29/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
The complex process known as epithelial to mesenchymal transition (EMT) plays a fundamental role in several biological settings, encompassing embryonic development, wound healing, and pathological conditions such as cancer and fibrosis. In recent years, a bulk of research has brought to light the key role of copper, a trace element with essential functions in cellular metabolism, cancer initiation and progression. Indeed, copper, besides functioning as cofactor of enzymes required for essential cellular processes, such as energy production and oxidation reactions, has emerged as an allosteric regulator of kinases whose activity is required to fulfill cancer dissemination through the EMT. In this comprehensive review, we try to describe the intricate relationship between the transition metal copper and EMT, spanning from the earliest foundational studies to the latest advancements. Our aim is to shed light on the multifaceted roles undertaken by copper in EMT in cancer and to unveil the diverse mechanisms by which copper homeostasis exerts its influence over EMT regulators, signaling pathways, cell metabolic reprogramming and transcription factors ultimately contributing to the spread of cancer. Therefore, this review not only may contribute to a deeper comprehension of copper-mediated mechanisms in EMT but also supports the hypothesis that targeting copper may contribute to counteract the progression of EMT-associated pathologies.
Collapse
Affiliation(s)
- Antonio Focaccio
- PhD School in Cellular and Molecular Biology, Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Luisa Rossi
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Anastasia De Luca
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy.
| |
Collapse
|
27
|
Wang J, Lv C, Wei X, Li F. Molecular mechanisms and therapeutic strategies for ferroptosis and cuproptosis in ischemic stroke. Brain Behav Immun Health 2024; 40:100837. [PMID: 39228970 PMCID: PMC11369453 DOI: 10.1016/j.bbih.2024.100837] [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: 01/15/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 09/05/2024] Open
Abstract
Ischemic stroke, as one of the most severe and prevalent neurological disorders, poses a significant threat to the health and quality of life of affected individuals. Stemming from the obstruction of blood flow, ischemic stroke, leads to cerebral tissue hypoxia and ischemia, instigating a cascade of pathophysiological changes that markedly exacerbate neuronal damage and may even culminate in cell death. In recent years, emerging research has increasingly focused on novel cell death mechanisms such as ferroptosis and cuproptosis. Mounting evidence underscores the independent roles of ferroptosis and cuproptosis in ischemic stroke. This review aims to elucidate potential cross-regulatory mechanisms between ferroptosis and cuproptosis, exploring their regulatory roles in ischemic stroke. The objective is to provide targeted therapeutic intervention strategies.
Collapse
Affiliation(s)
- Jing Wang
- Department of neurology, Lu 'an Municipal People's Hospital, Anhui, China
- Bengbu Medical College, Anhui, China
| | - Cunming Lv
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Xinyu Wei
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Feng Li
- Department of neurology, Lu 'an Municipal People's Hospital, Anhui, China
| |
Collapse
|
28
|
Basaran B, Turk H. The levels, single and multiple health risk assessment of 23 metals in enteral nutrition formulas. Food Chem Toxicol 2024; 192:114914. [PMID: 39127122 DOI: 10.1016/j.fct.2024.114914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Enteral nutrition formulas are products that provide macro and micronutrients to patients who cannot receive their nutrition orally. In this study, the levels of 23 metals known to have potential health risks were determined by inductively coupled plasma mass spectrometry in a total of 28 enteral nutrition formula. Metal exposure was calculated according to three different daily energy intake scenarios (Scenario 1 = 50% oral nutrition + 50% enteral nutrition formula, Scenario 2 = 25% oral nutrition + 75% enteral nutrition formula and Scenario 3 = 100% enteral nutrition formula) and evaluated in terms of non-carcinogenic health risks. The mean levels of Fe, Co, Ni, Cu, Zn, Mo, Se, Li, Be, V, As, Sr, Ag, Cd, Sb, Ba, La, Hg and Pb in the samples analyzed were determined 12,000 ± 3300, 64 ± 1.6, 10 ± 13, 1300 ± 400, 8500 ± 2500, 75 ± 30, 61 ± 21, 0.34 ± 0.36, 0.05 ± 0.08, 7.3 ± 2, 1.6 ± 0.6, 457 ± 166, 0.02 ± 0.1, 0.14 ± 0.12, 0.01 ± 0.1, 74 ± 103, 0.63 ± 0.4, 0.05 ± 0.03 and 0.14 ± 0.7 μg/L. These metals were considered safe in terms of non-carcinogenic health risks when analyzed individually. However, when the target hazard quotient values of all metals were evaluated together, hazard index values were higher than the reference value of 1, for both men and women, indicating potential health risks.
Collapse
Affiliation(s)
- Burhan Basaran
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Recep Tayyip Erdogan University, Rize, 53100, Türkiye.
| | - Hulya Turk
- Department of Biology, Science Faculty, Ataturk University, Erzurum, 25240, Türkiye
| |
Collapse
|
29
|
Yang Y, Wu J, Wang L, Ji G, Dang Y. Copper homeostasis and cuproptosis in health and disease. MedComm (Beijing) 2024; 5:e724. [PMID: 39290254 PMCID: PMC11406047 DOI: 10.1002/mco2.724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Copper is a vital trace element in human physiology, essential for the synthesis of numerous crucial metabolic enzymes and facilitation of various biological processes. Regulation of copper levels within a narrow range is imperative for maintaining metabolic homeostasis. Numerous studies have demonstrated the significant roles of copper homeostasis and cuproptosis in health and disease pathogenesis. However, a comprehensive and up-to-date systematic review in this domain remains absent. This review aims to consolidate recent advancements in understanding the roles of cuproptosis and copper homeostasis in health and disease, focusing on the underlying mechanisms and potential therapeutic interventions. Dysregulation of copper homeostasis, manifesting as either copper excess or deficiency, is implicated in the etiology of various diseases. Cuproptosis, a recently identified form of cell death, is characterized by intracellular copper overload. This phenomenon mediates a diverse array of evolutionary processes in organisms, spanning from health to disease, and is implicated in genetic disorders, liver diseases, neurodegenerative disorders, and various cancers. This review provides a comprehensive summary of the pathogenic mechanisms underlying cuproptosis and copper homeostasis, along with associated targeted therapeutic agents. Furthermore, it explores future research directions with the potential to yield significant advancements in disease treatment, health management, and disease prevention.
Collapse
Affiliation(s)
- Yunuo Yang
- Institute of Digestive Diseases China-Canada Center of Research for Digestive Diseases Longhua Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine) Shanghai China
| | - Jiaxuan Wu
- Institute of Digestive Diseases China-Canada Center of Research for Digestive Diseases Longhua Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine) Shanghai China
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine University of Ottawa Ottawa Ontario Canada
- China-Canada Centre of Research for Digestive Diseases University of Ottawa Ottawa Ontario Canada
| | - Guang Ji
- Institute of Digestive Diseases China-Canada Center of Research for Digestive Diseases Longhua Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine) Shanghai China
| | - Yanqi Dang
- Institute of Digestive Diseases China-Canada Center of Research for Digestive Diseases Longhua Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine) Shanghai China
| |
Collapse
|
30
|
Weishaupt AK, Ruecker L, Meiners T, Schwerdtle T, Silva Avila D, Aschner M, Bornhorst J. Copper-mediated neurotoxicity and genetic vulnerability in the background of neurodegenerative diseases in C. elegans. Toxicol Sci 2024; 201:254-262. [PMID: 39067045 PMCID: PMC11424883 DOI: 10.1093/toxsci/kfae092] [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] [Indexed: 07/30/2024] Open
Abstract
The mechanisms associated with neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), have yet to be fully characterized, and genetic as well as environmental factors in their disease etiology are underappreciated. Although mutations in genes such as PARKIN and LRRK2 have been linked to PD, the idiopathic component of the disease suggests a contribution of environmental risk factors, including metals, such as copper (Cu). Cu overexposure has been reported to cause oxidative stress and neurotoxicity, but its role in neurodegenerative diseases is rarely studied. Using Caenorhabditis elegans (C. elegans) as a model organism for neurotoxicity, we assessed the effects of Cu oversupply in AD and PD models. Our findings reveal that although copper treatment did not induce neurodegeneration in wild-type worms or the AD model, it significantly exacerbated neurodegeneration in the PD-associated mutants PARKIN and LRRK2. These results suggest that genetic predisposition for PD enhances the sensitivity to copper toxicity, highlighting the multifactorial nature of neurodegenerative diseases. Furthermore, our study provides insight into the mechanisms underlying Cu-induced neurotoxicity in PD models, including disruptions in dopamine levels, altered dopamine-dependent behavior and degraded dopaminergic neurons. Overall, our novel findings contribute to a better understanding of the complex interactions between genetic susceptibility, environmental factors, and neurodegenerative disease pathogenesis, emphasizing the importance of a tightly regulated Cu homeostasis in the etiology of PD. Copper oversupply exacerbated neurodegeneration in Caenorhabditis elegans models of Parkinson's disease, highlighting the genetic susceptibility and emphasizing the crucial role of tightly regulated copper homeostasis in Parkinson's disease pathogenesis.
Collapse
Affiliation(s)
- Ann-Kathrin Weishaupt
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Potsdam, Jena, Wuppertal, Germany
| | - Lysann Ruecker
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
| | - Torben Meiners
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
| | - Tanja Schwerdtle
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Potsdam, Jena, Wuppertal, Germany
- German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Daiana Silva Avila
- Laboratory of Toxicology and Biochemistry in Caenorhabditis elegans, Universidade Federal do Pampa, 97501-970 Uruguaiana, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Julia Bornhorst
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Potsdam, Jena, Wuppertal, Germany
| |
Collapse
|
31
|
Huang Q, Li J, Qi Y, He X, Shen C, Wang C, Wang X, Xia Q, Zhang Y, Pan Z, Hu Q, Cao Z, Liu Y, Huang J, Han G, Zheng Y, Zheng B, Zeng X, Bi X, Yu J. Copper overload exacerbates testicular aging mediated by lncRNA:CR43306 deficiency through ferroptosis in Drosophila. Redox Biol 2024; 76:103315. [PMID: 39154546 PMCID: PMC11378248 DOI: 10.1016/j.redox.2024.103315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024] Open
Abstract
Testicular aging manifests as impaired spermatogenesis and morphological alterations in Drosophila. Nonetheless, the comprehensive molecular regulatory framework remains largely undisclosed. This investigation illustrates the impact of copper overload on testicular aging and underscores the interplay between copper overload and lncRNA. Copper overload triggers Cuproptosis through the mitochondrial TCA cycle, facilitating intracellular interactions with Ferroptosis, thereby governing testicular aging. Dysfunction of lncRNA:CR43306 also contributes to testicular aging in Drosophila, emphasizing the significance of lncRNA:CR43306 as a novel aging-associated lncRNA. Moreover, copper overload exacerbates spermatid differentiation defects mediated by lncRNA:CR43306 deficiency through oxidative stress, copper, and iron transport. Therapeutically, Ferrostatin-1 and Resveratrol emerge as potential remedies for addressing testicular aging. This study offers perspectives on the regulatory mechanisms involving copper overload and lncRNA:CR43306 deficiency in the context of testicular aging.
Collapse
Affiliation(s)
- Qiuru Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Jiaxin Li
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Yujuan Qi
- Clinical Center of Reproductive Medicine, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, 221000, China
| | - Xuxin He
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Cong Shen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, 215002, China
| | - Chenyu Wang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Xinda Wang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Qiushi Xia
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Yi Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Ziyue Pan
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Qingqing Hu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Ziyu Cao
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Yiheng Liu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Jingqi Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Guoqing Han
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Ying Zheng
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Bo Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, 215002, China.
| | - Xuhui Zeng
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Xiaolin Bi
- School of Medicine, Nantong University, Nantong, 226001, China.
| | - Jun Yu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| |
Collapse
|
32
|
Kamel AY, Johnson ZD, Hernandez I, Nguyen C, Rolfe M, Joseph T, Dixit D, Shen S, Chaudhry N, Pham A, Rampertab SD, Zimmermann E. Micronutrient deficiencies in inflammatory bowel disease: an incidence analysis. Eur J Gastroenterol Hepatol 2024; 36:1186-1192. [PMID: 39012640 DOI: 10.1097/meg.0000000000002821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
BACKGROUND Micronutrient deficiencies associated with malnutrition in patients with inflammatory bowel disease (IBD) can lead to complications including anemia, coagulopathy, poor wound healing, and colorectal cancer. This study aimed to investigate micronutrient deficiencies (copper, vitamins A, B 9 , E, and K) in IBD patients and highlight associated symptoms to aid in the recognition of micronutrient deficiencies. METHODS A retrospective electronic chart review was performed on adults diagnosed with Crohn's disease or ulcerative colitis hospitalized at a tertiary care center for IBD flare between January 2013 and June 2017. Patients with serum or whole blood micronutrient levels were included. Pregnant and incarcerated patients were excluded. RESULTS A total of 611 IBD patients (440 Crohn's disease, 171 ulcerative colitis) met the inclusion criteria. Micronutrients were assessed in a subset of IBD patients (copper: 12.3%, A: 10.1%, B 9 : 95.9%, E: 10.3%, and K: 4.6%). Overall, 10.1% of patients had micronutrient deficiencies. The proportion of patients with copper, A, B 9 , E, and K deficiencies were 25.4, 53.3, 1.9, 23.7, and 29.4% for Crohn's disease and 50, 52.9, 1.2, 43.8, and 18.2% for ulcerative colitis, respectively. The most common symptoms or historical features associated with micronutrient deficiency were anemia (copper, B 9 ), muscle weakness (copper, E) thrombocytopenia, fatigue (copper, B 9 ), diarrhea (B 9 ), dry skin, hyperkeratosis, pruritus, significant weight loss, elevated C-reactive protein (A), bleeding, and osteoporosis (K). CONCLUSION Micronutrient deficiencies are common in IBD patients, yet they are not routinely assessed. Copper, vitamins A, E, and K deficiencies are particularly underrecognized. Associated historical features should raise suspicion and prompt assessment and treatment.
Collapse
Affiliation(s)
- Amir Y Kamel
- Department of Pharmacy, UF Health Shands Hospital
| | | | - Isabela Hernandez
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville
| | | | - Melanie Rolfe
- Department of Pharmacy Services, Lee Health, Fort Myers
| | - Tiffany Joseph
- Department of Pharmacy, Orlando Health Orlando Regional Medical Center, Orlando
| | - Devika Dixit
- Department of Internal Medicine, University of Florida, Gainesville, Florida, USA
| | - Steve Shen
- Department of Internal Medicine, University of Florida, Gainesville, Florida, USA
| | - Naueen Chaudhry
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville
| | - Angela Pham
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville
| | - S Devi Rampertab
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville
| | - Ellen Zimmermann
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville
| |
Collapse
|
33
|
Alanazi ST, Salama SA, Althobaiti MM, Alotaibi RA, AlAbdullatif AA, Musa A, Harisa GI. Alleviation of Copper-Induced Hepatotoxicity by Bergenin: Diminution of Oxidative Stress, Inflammation, and Apoptosis via Targeting SIRT1/FOXO3a/NF-κB Axes and p38 MAPK Signaling. Biol Trace Elem Res 2024:10.1007/s12011-024-04401-3. [PMID: 39347884 DOI: 10.1007/s12011-024-04401-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Despite its biological importance, excess copper induces organ damage, especially to the liver. Disruption of critical signaling cascades that control redox status, inflammatory responses, and cellular apoptosis significantly contributes to the copper-induced hepatotoxicity. The present work explored the hepatoprotective ability of bergenin against the copper-induced hepatotoxicity using male Wistar rats as a mammalian model. The results revealed that bergenin suppressed the copper-evoked histopathological changes and hepatocellular necrosis as indicated by decreased activity of the liver enzymes ALT and AST in the sera of the copper-intoxicated rats. It decreased hepatic copper content and the copper-induced oxidative stress as revealed by reduced lipid peroxidation and improved activity of the antioxidant enzymes thioredoxin reductase, glutathione peroxidase, catalase, and superoxide dismutase. Bergenin downregulated the inflammatory cytokines TNF-α and IL-6, and the inflammatory cell infiltration to the liver tissues. Additionally, it inhibited the copper-induced apoptosis as indicated by significant reduction in caspase-3 activity. At the molecular level, bergenin activated the antioxidant transcription factor FOXO3a, inhibited the nuclear translocation of the inflammatory transcription factor NF-κB, and suppressed the inflammatory signaling molecules p38 MAPK and c-Fos. Interestingly, bergenin improved the expression of the anti-apoptotic protein Bcl2 and reduced the pro-apoptotic protein BAX. Bergenin markedly enhanced the expression of the histone deacetylase protein SIRT1 that regulates activity of NF-κB and FOXO3a. Collectively, these findings highlight the alleviating activity of bergenin against the copper-induced hepatotoxicity via controlling oxidative stress, inflammation, and apoptosis potentially through upregulation of SIRT1, activation of FOXO3a along with suppression of NF-κB and p38 MAPK signaling.
Collapse
Affiliation(s)
- Samyah T Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, 11433, Riyadh, Saudi Arabia
| | - Samir A Salama
- Division of Biochemistry, Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia.
| | - Musaad M Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Rana A Alotaibi
- College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Ammar A AlAbdullatif
- Pharmaceutical Care Services, Ministry of the National Guard-Health Affairs, P.O. Box 4616, 31412, Dammam, Saudi Arabia
| | - Arafa Musa
- Department of Pharmacognosy, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Gamaleldin I Harisa
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
34
|
Peng G, Huang Y, Xie G, Tang J. Exploring Copper's role in stroke: progress and treatment approaches. Front Pharmacol 2024; 15:1409317. [PMID: 39391696 PMCID: PMC11464477 DOI: 10.3389/fphar.2024.1409317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Copper is an important mineral, and moderate copper is required to maintain physiological processes in nervous system including cerebral ischemia/reperfusion (I/R) injury. Over the past few decades, copper induced cell death, named cuprotosis, has attracted increasing attention. Several lines of evidence have confirmed cuprotosis exerts pivotal role in diverse of pathological processes, such as cancer, neurodegenerative diseases, and I/R injury. Therefore, an in-depth understanding of the interaction mechanism between copper-mediated cell death and I/R injury may reveal the significant alterations about cellular copper-mediated homeostasis in physiological and pathophysiological conditions, as well as therapeutic strategies deciphering copper-induced cell death in cerebral I/R injury.
Collapse
Affiliation(s)
- Gang Peng
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan, China
| | - Yongpan Huang
- School of Medicine, Changsha Social Work College, Changsha, Hunan, China
| | - Guangdi Xie
- Department of Neurology, Huitong People’s Hospital, Huitong, Hunan, China
| | - Jiayu Tang
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
35
|
Rodriguez P, Kalia V, Fenollar-Ferrer C, Gibson CL, Gichi Z, Rajoo A, Matier CD, Pezacki AT, Xiao T, Carvelli L, Chang CJ, Miller GW, Khamoui AV, Boerner J, Blakely RD. Glial swip-10 controls systemic mitochondrial function, oxidative stress, and neuronal viability via copper ion homeostasis. Proc Natl Acad Sci U S A 2024; 121:e2320611121. [PMID: 39288174 PMCID: PMC11441482 DOI: 10.1073/pnas.2320611121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/01/2024] [Indexed: 09/19/2024] Open
Abstract
Cuprous copper [Cu(I)] is an essential cofactor for enzymes that support many fundamental cellular functions including mitochondrial respiration and suppression of oxidative stress. Neurons are particularly reliant on mitochondrial production of ATP, with many neurodegenerative diseases, including Parkinson's disease, associated with diminished mitochondrial function. The gene MBLAC1 encodes a ribonuclease that targets pre-mRNA of replication-dependent histones, proteins recently found in yeast to reduce Cu(II) to Cu(I), and when mutated disrupt ATP production, elevates oxidative stress, and severely impacts cell growth. Whether this process supports neuronal and/or systemic physiology in higher eukaryotes is unknown. Previously, we identified swip-10, the putative Caenorhabditis elegans ortholog of MBLAC1, establishing a role for glial swip-10 in limiting dopamine (DA) neuron excitability and sustaining DA neuron viability. Here, we provide evidence from computational modeling that SWIP-10 protein structure mirrors that of MBLAC1 and locates a loss of function coding mutation at a site expected to disrupt histone RNA hydrolysis. Moreover, we find through genetic, biochemical, and pharmacological studies that deletion of swip-10 in worms negatively impacts systemic Cu(I) levels, leading to deficits in mitochondrial respiration and ATP production, increased oxidative stress, and neurodegeneration. These phenotypes can be offset in swip-10 mutants by the Cu(I) enhancing molecule elesclomol and through glial expression of wildtype swip-10. Together, these studies reveal a glial-expressed pathway that supports systemic mitochondrial function and neuronal health via regulation of Cu(I) homeostasis, a mechanism that may lend itself to therapeutic strategies to treat devastating neurodegenerative diseases.
Collapse
Affiliation(s)
- Peter Rodriguez
- Department of Biological Sciences, Charles E. Schmidt College of Science, Boca Raton, FL33412
| | - Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032
| | - Cristina Fenollar-Ferrer
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, Bethesda, MD20892
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
| | - Chelsea L. Gibson
- Department of Biological Sciences, Charles E. Schmidt College of Science, Boca Raton, FL33412
- Oak Ridge Institute for Science and Education, Oak Ridge, TN37830
| | - Zayna Gichi
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL33458
| | - Andre Rajoo
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL33458
| | - Carson D. Matier
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
| | - Aidan T. Pezacki
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Princeton University, Princeton, NJ08544
| | - Tong Xiao
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Princeton University, Princeton, NJ08544
| | - Lucia Carvelli
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL33458
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL33458
| | - Christopher J. Chang
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Princeton University, Princeton, NJ08544
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032
| | - Andy V. Khamoui
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
- Department of Exercise Science and Health Promotion, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL33431
| | - Jana Boerner
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
| | - Randy D. Blakely
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL33458
| |
Collapse
|
36
|
Qin L, Cao X, Huang T, Liu Y, Li S. Identification of potential biomarkers of cuproptosis in cerebral ischemia. Front Nutr 2024; 11:1410431. [PMID: 39360273 PMCID: PMC11445069 DOI: 10.3389/fnut.2024.1410431] [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: 04/01/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Objective Cerebral ischemia can cause mild damage to local brain nerves due to hypoxia and even lead to irreversible damage due to neuronal cell death. However, the underlying pathogenesis of this phenomenon remains unclear. This study utilized bioinformatics to explore the role of cuproptosis in cerebral ischemic disease and its associated biomarkers. Method R software identified the overlap of cerebral ischemia and cuproptosis genes, analyzed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), and explored hub genes. Expressions and localizations of hub genes in brain tissue, cells, and immune cells were analyzed, along with predictions of protein structures, miRNAs, and transcription factors. A network was constructed depicting hub gene co-expression with miRNAs and interactions with transcription factors. Ferredoxin 1 (FDX1) expression was determined using qRT-PCR. Results Ten cuproptosis-related genes in cerebral ischemia were identified, with GO analysis revealing involvement in acetyl-CoA synthesis, metabolism, mitochondrial function, and iron-sulfur cluster binding. KEGG highlighted processes like the tricarboxylic acid cycle, pyruvate metabolism, and glycolysis/gluconeogenesis. Using the Human Protein Atlas, eight hub genes associated with cuproptosis were verified in brain tissues, hippocampus, and AF22 cells. Lipoyl(octanoyl) transferase 1 (LIPT1), was undetected, while others were found in mitochondria or both nucleus and mitochondria. These genes were differentially expressed in immune cells. FDX1, lipoic acid synthetase (LIAS), dihydrolipoamide S-acetyltransferase (DLAT), pyruvate dehydrogenase E1 component subunit alpha 1 (PDHA1), PDHB, and glutaminase (GLS) were predicted to target 111 miRNAs. PDHA1, FDX1, LIPT1, PDHB, LIAS, DLAT, GLS, and dihydrolipoamide dehydrogenase (DLD) were predicted to interact with 11, 10, 10, 9, 8, 7, 5, and 4 transcription factors, respectively. Finally, FDX1 expression was significantly upregulated in the hippocampus of ovariectomized rats with ischemia. Conclusion This study revealed an association between cerebral ischemic disease and cuproptosis, identifying eight potential target genes. These findings offer new insights into potential biomarkers for the diagnosis, treatment, and prognosis of cerebral ischemia, and provide avenues for the exploration of new medical intervention targets.
Collapse
Affiliation(s)
- Lihua Qin
- School of Nursing, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardiocerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xi Cao
- School of Nursing, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tengjia Huang
- School of Nursing, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yixin Liu
- School of Nursing, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Sheng Li
- Key Laboratory of Hunan Province for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardiocerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
37
|
Yu Q, Xiao Y, Guan M, Zhang X, Yu J, Han M, Li Z. Copper metabolism in osteoarthritis and its relation to oxidative stress and ferroptosis in chondrocytes. Front Mol Biosci 2024; 11:1472492. [PMID: 39329090 PMCID: PMC11425083 DOI: 10.3389/fmolb.2024.1472492] [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: 07/29/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Ferroptosis, an iron-ion-dependent process of lipid peroxidation, damages the plasma membrane, leading to non-programmed cell death. Osteoarthritis (OA), a prevalent chronic degenerative joint disease among middle-aged and older adults, is characterized by chondrocyte damage or loss. Emerging evidence indicates that chondrocyte ferroptosis plays a role in OA development. However, most research has concentrated on ferroptosis regulation involving typical iron ions, potentially neglecting the significance of elevated copper ions in both serum and joint fluid of patients with OA. This review aims to fill this gap by systematically examining the interplay between copper metabolism, oxidative stress, ferroptosis, and copper-associated cell death in OA. It will provide a comprehensive overview of copper ions' role in regulating ferroptosis and their dual role in OA. This approach seeks to offer new insights for further research, prevention, and treatment of OA.
Collapse
Affiliation(s)
- Qingyuan Yu
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Yanan Xiao
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Mengqi Guan
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xianshuai Zhang
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jianan Yu
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Mingze Han
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Zhenhua Li
- Orthopedic Center, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| |
Collapse
|
38
|
Zu H, Wu Y, Meng H, Cheng X, Wang Y, Zhang LW, Wang Y. Tumor Metabolism Aiming Cu 2-xS Nanoagents Mediate Photothermal-Derived Cuproptosis and Immune Activation. ACS NANO 2024; 18:23941-23957. [PMID: 39171773 DOI: 10.1021/acsnano.3c10588] [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: 08/23/2024]
Abstract
Cuproptosis is an emerging form of cell death that relies on the targeted delivery of copper ions to lipoylated tricarboxylic acid cycle proteins. However, a major challenge associated with cuproptosis is its potential to kill both normal and tumor cells without discrimination. Therefore, it is crucial to develop strategies for precise intracellular delivery and redox control of copper to create effective cuproptosis-based tumor therapies. We have introduced a class of nanoagents called metabolism aiming Cu2-xS (MACuS) through a glucose-mediated biomineralization approach. MACuS nanoagents can be specifically targeted to tumors via the glucose transport receptor 1, and we found that NIR-II irradiation can not only result in direct hyperthermia ablation of tumor cells but also facilitate efficient cuproptosis and enhance reactive oxygen species-induced cytotoxicity in tumor cells. As a result, the triple effect of MACuS treatment induced immunogenic cell death, which triggered systemic antitumor immune responses and demonstrated potent efficacy in inhibiting growth, metastasis, and recurrence in mouse and rabbit breast cancer models. The precise intracellular delivery and redox control of copper provided by MACuS hold great potential for the development of highly efficient cuproptosis-based tumor therapies with minimal off-target effects.
Collapse
Affiliation(s)
- He Zu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yanxian Wu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Hezhang Meng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xiaju Cheng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yangyun Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Leshuai W Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yong Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
39
|
Sailer J, Nagel J, Akdogan B, Jauch AT, Engler J, Knolle PA, Zischka H. Deadly excess copper. Redox Biol 2024; 75:103256. [PMID: 38959622 PMCID: PMC11269798 DOI: 10.1016/j.redox.2024.103256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024] Open
Abstract
Higher eukaryotes' life is impossible without copper redox activity and, literally, every breath we take biochemically demonstrates this. However, this dependence comes at a considerable price to ensure target-oriented copper action. Thereto its uptake, distribution but also excretion are executed by specialized proteins with high affinity for the transition metal. Consequently, malfunction of copper enzymes/transporters, as is the case in hereditary Wilson disease that affects the intracellular copper transporter ATP7B, comes with serious cellular damage. One hallmark of this disease is the progressive copper accumulation, primarily in liver but also brain that becomes deadly if left untreated. Such excess copper toxicity may also result from accidental ingestion or attempted suicide. Recent research has shed new light into the cell-toxic mechanisms and primarily affected intracellular targets and processes of such excess copper that may even be exploited with respect to cancer therapy. Moreover, new therapies are currently under development to fight against deadly toxic copper.
Collapse
Affiliation(s)
- Judith Sailer
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Judith Nagel
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Banu Akdogan
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Adrian T Jauch
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Jonas Engler
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Percy A Knolle
- Institute of Molecular Immunology and Experimental Oncology, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Hans Zischka
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine and Health, Munich, Germany; Institute of Molecular Toxicology and Pharmacology, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
40
|
Hao D, Luo W, Yan Y, Zhou J. Focus on cuproptosis: Exploring new mechanisms and therapeutic application prospects of cuproptosis regulation. Biomed Pharmacother 2024; 178:117182. [PMID: 39053428 DOI: 10.1016/j.biopha.2024.117182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
Cuproptosis is a novel form of regulated cell death, which plays an important role in the physiological and pathological processes of the human body. Despite the increasing research on cuproptosis-related genes (CRGs) and their correlation with diseases, the pathogenesis of cuproptosis-related diseases remains unclear. Furthermore, there is a lack of reviews on the emerging technologies for regulating cuproptosis in disease treatment. This study delves into the copper-induced cell death mechanism, distinguishing cuproptosis from mechanisms like oxidative stress, glutathione synthesis inhibition, and ubiquitin-proteasome system inhibition. Several long-standing mysteries of diseases such as Wilson's disease and Menkes disease may be attributed to the occurrence of cuproptosis. In addition, we also review the detection indicators related to cuproptosis, providing targets for the diagnosis of cuproptosis-related diseases, and summarize the application value of cuproptosis in tumor therapy to better elucidate the impact of copper in cell death and diseases, and thus to promote the application prospects and possible strategies of cuproptosis-related substances, such as copper ion chelators, copper ion carriers, and copper nanomaterials, in disease therapy.
Collapse
Affiliation(s)
- Donglin Hao
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China; Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Wei Luo
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China; Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China
| | - Yongmin Yan
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China; Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China.
| | - Jing Zhou
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China; Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou, China.
| |
Collapse
|
41
|
Qiao L, Xuan W, Ou Y, Li L, Wu S, Guo Y, Liu M, Yu D, Chen Q, Yuan J, Zuo M, Wei C, Ou C, Li H, Cheng D, Yu Z, Li Z. Tumor microenvironment activation amplify oxidative stress promoting tumor energy remodeling for mild photothermal therapy and cuproptosis. Redox Biol 2024; 75:103260. [PMID: 38955114 PMCID: PMC11267038 DOI: 10.1016/j.redox.2024.103260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
Tumor metabolic reprogramming requires high levels of adenosine triphosphate (ATP) to maintain treatment resistance, which poses major challenges to chemotherapy and photothermal therapy. Especially, high levels of ATP promote copper ion efflux for limiting the curative effect of cuproptosis. Here, an H2S-responsive mesoporous Cu2Cl(OH)3-loading chemotherapeutic cisplatin (CDDP) was synthesized, and the final nanoparticle, CDDP@Cu2Cl(OH)3-CDs (CDCuCDs), was encapsulated by electrostatic action with carbon dots (CDs). CDCuCDs reacted with overproduction H2S in colon tumor to produce photothermic copper sulfide for photothermal therapy. CDDP was released by lysis to achieve chemotherapeutic effects. Importantly, CDDP elevated H2O2 levels in cells through a cascade reaction and continuously transforms H2O2 into highly cytotoxic •OH through chemodynamic therapy between H2O2 and Cu+, which enables nanoparticles to generate •OH and improve the chemotherapeutic efficacy. Highly toxic •OH disrupts mitochondrial homeostasis, prohibiting it from performing normal energy-supplying functions. Down-regulated ATP inhibits heat shock protein expression, which promotes the therapeutic effect of mild photothermal therapy and reduces the efflux of intracellular copper ions, thus improving the therapeutic effect of cuproptosis. Our research provides a potential therapeutic strategy using overproduction H2S responses in tumors, allowing tumor microenvironment-activated •OH nanogenerators to promote tumor energy remodeling for cancer treatment.
Collapse
Affiliation(s)
- Lihong Qiao
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, People's Republic of China; Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, People's Republic of China; Department of Laboratory Medicine Dongguan Institute of Clinical Cancer Research the Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, People's Republic of China; Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Wenting Xuan
- Department of Endocrinology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, People's Republic of China
| | - Yijing Ou
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, People's Republic of China; Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, People's Republic of China
| | - Lin Li
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, People's Republic of China; Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, People's Republic of China
| | - Shuzhen Wu
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, People's Republic of China; Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, People's Republic of China
| | - Yanxian Guo
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, People's Republic of China; Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, People's Republic of China
| | - Mu Liu
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, People's Republic of China; Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, People's Republic of China
| | - Dongsheng Yu
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Qinghua Chen
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Jianmin Yuan
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Mingxiang Zuo
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Chuanqi Wei
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Chiyi Ou
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Haowen Li
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Du Cheng
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China.
| | - Zhiqiang Yu
- Department of Laboratory Medicine Dongguan Institute of Clinical Cancer Research the Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, People's Republic of China.
| | - Zhongjun Li
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, People's Republic of China; Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, 523058, People's Republic of China.
| |
Collapse
|
42
|
Wróblewski M, Wróblewska W, Sobiesiak M. The Role of Selected Elements in Oxidative Stress Protection: Key to Healthy Fertility and Reproduction. Int J Mol Sci 2024; 25:9409. [PMID: 39273356 PMCID: PMC11395468 DOI: 10.3390/ijms25179409] [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: 08/03/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Oxidative stress and its relationship to fertility and reproduction is a topic of interest in medicine, especially in the context of the effects of trace elements and micronutrients. Oxidative stress occurs when there is an excess of free radicals in the body, which can lead to cell and tissue damage. Free radicals are reactive oxygen species (ROS) that can be formed as a result of normal metabolic processes, as well as under the influence of external factors such as environmental pollution, UV radiation, and diet. Oxidative stress has a significant impact on fertility. In men, it can lead to DNA damage in sperm, which can result in reduced semen quality, reduced sperm motility and increased numbers of defective sperm, and free radical damage to sperm cell membranes causing a reduction in the number of available sperm. In women, oxidative stress can affect the quality of female reproductive cells, which can lead to problems with their maturation and with embryo implantation in the uterus and can also affect ovarian function and disrupt hormonal regulation of the menstrual cycle. A proper balance of trace elements and micronutrients is key to protecting against oxidative stress and maintaining reproductive health. Supplementation with appropriate elements such as zinc, selenium, copper, manganese, chromium, and iron can help reduce oxidative stress and improve fertility. This work discusses the effects of selected elements on oxidative stress parameters specifically in terms of fertility and reproduction.
Collapse
Affiliation(s)
- Marcin Wróblewski
- Department of Medical Biology and Biochemistry, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Weronika Wróblewska
- Student Research Club of Medical Biology and Biochemistry, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Marta Sobiesiak
- Department of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| |
Collapse
|
43
|
Matysik J, Długosz O, Banach M. Development of Nanozymatic Characteristics in Metal-Doped Oxide Nanomaterials. J Phys Chem B 2024; 128:8007-8016. [PMID: 39120940 PMCID: PMC11345814 DOI: 10.1021/acs.jpcb.4c02526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
Nanozymes are nanoscale materials that exhibit enzymatic-like activity, combining the benefits of nanomaterials with biocatalytic effects. The addition of metals to nanomaterials can enhance their nanozyme activity by mimicking the active sites of enzymes, providing structural support and promoting redox activity. In this study, nanostructured oxide and silicate-phosphate nanomaterials with varying manganese and copper additions were characterized. The objective was to assess the influence of metal modifications (Mn and Cu) on the acquisition of the nanozymatic activity by selected nanomaterials. An increase in manganese content in each material enhanced proteolytic activity (from 20 to 40 mUnit/mg for BG-Mn), while higher copper addition in glassy materials increased activity by 40%. Glassy materials exhibited approximately twice the 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid radical activity (around 40 μmol/mL) compared to that of oxide materials. The proteolytic and antioxidant activities discussed in the study can be considered indicators for evaluating the enzymatic properties of the nanomaterials. Observations conducted on nanomaterials may aid in the development of materials with enhanced catalytic efficiency and a wide range of applications.
Collapse
Affiliation(s)
- Julia Matysik
- Faculty of Chemical Engineering and Technology, Institute of Chemistry and Inorganic Technology, Cracow University of Technology, Warszawska St. 24, Cracow 31-155, Poland
| | - Olga Długosz
- Faculty of Chemical Engineering and Technology, Institute of Chemistry and Inorganic Technology, Cracow University of Technology, Warszawska St. 24, Cracow 31-155, Poland
| | - Marcin Banach
- Faculty of Chemical Engineering and Technology, Institute of Chemistry and Inorganic Technology, Cracow University of Technology, Warszawska St. 24, Cracow 31-155, Poland
| |
Collapse
|
44
|
Charkiewicz AE. Is Copper Still Safe for Us? What Do We Know and What Are the Latest Literature Statements? Curr Issues Mol Biol 2024; 46:8441-8463. [PMID: 39194715 DOI: 10.3390/cimb46080498] [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: 06/26/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Copper (Cu) is a precious metal and one of the three most abundant trace elements in the body (50-120 mg). It is involved in a large number of cellular mechanisms and pathways and is an essential cofactor in the function of cellular enzymes. Both its excess and deficiency may be harmful for many diseases. Even small changes in Cu concentration may be associated with significant toxicity. Consequently, it can be damaging to any organ or tissue in our body, beginning with harmful effects already at the molecular level and then affecting the degradation of individual tissues/organs and the slow development of many diseases, such as those of the immunological system, skeletal system, circulatory system, nervous system, digestive system, respiratory system, reproductive system, and skin. The main purpose of this article is to review the literature with regard to both the healthiness and toxicity of copper to the human body. A secondary objective is to show its widespread use and sources, including in food and common materials in contact with humans. Its biological half-life from diet is estimated to range from 13 to 33 days. The retention or bioavailability of copper from the diet is influenced by several factors, such as age, amount and form of copper in the diet, lifestyle, and genetic background. The upper limit of normal in serum in healthy adults is approximately 1.5 mg Cu/L, while the safe upper limit of average intake is set at 10-12 mg/day, the reference limit at 0.9 mg/day, and the minimum limit at 0.6-0.7 mg/day. Cu is essential, and in the optimal dose, it provides antioxidant defense, while its deficiency reduces the body's ability to cope with oxidative stress. The development of civilization and the constant, widespread use of Cu in all electrical devices will not be stopped, but the health of people directly related to its extraction, production, or distribution can be controlled, and the inhabitants of nearby towns can be protected. It is extremely difficult to assess the effects of copper on the human body because of its ubiquity and the increasing reports in the literature about its effects, including copper nanoparticles.
Collapse
|
45
|
Tetteh PA, Kalvani Z, Stevens D, Sappal R, Kamunde C. Interactions of binary mixtures of metals on rainbow trout (Oncorhynchus mykiss) heart mitochondrial H 2O 2 homeodynamics. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 273:106986. [PMID: 38851027 DOI: 10.1016/j.aquatox.2024.106986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/10/2024]
Abstract
For continuous pumping of blood, the heart needs a constant supply of energy (ATP) that is primarily met via oxidative phosphorylation in the mitochondria of cardiomyocytes. However, sustained high rates of electron transport for energy conversion redox reactions predisposes the heart to the production of reactive oxygen species (ROS) and oxidative stress. Mitochondrial ROS are fundamental drivers of responses to environmental stressors including metals but knowledge of how combinations of metals alter mitochondrial ROS homeodynamics remains sparse. We explored the effects and interactions of binary mixtures of copper (Cu), cadmium (Cd), and zinc (Zn), metals that are common contaminants of aquatic systems, on ROS (hydrogen peroxide, H2O2) homeodynamics in rainbow trout (Oncorhynchus mykiss) heart mitochondria. Isolated mitochondria were energized with glutamate-malate or succinate and exposed to a range of concentrations of the metals singly and in equimolar binary concentrations. Speciation analysis revealed that Cu was highly complexed by glutamate or Tris resulting in Cu2+ concentrations in the picomolar to nanomolar range. The concentration of Cd2+ was 7.2-7.5 % of the total while Zn2+ was 15 % and 21 % of the total during glutamate-malate and succinate oxidation, respectively. The concentration-effect relationships for Cu and Cd on mitochondrial H2O2 emission depended on the substrate while those for Zn were similar during glutamate-malate and succinate oxidation. Cu + Zn and Cu + Cd mixtures exhibited antagonistic interactions wherein Cu reduced the effects of both Cd and Zn, suggesting that Cu can mitigate oxidative distress caused by Cd or Zn. Binary combinations of the metals acted additively to reduce the rate constant and increase the half-life of H2O2 consumption while concomitantly suppressing thioredoxin reductase and stimulating glutathione peroxidase activities. Collectively, our study indicates that binary mixtures of Cu, Zn, and Cd act additively or antagonistically to modulate H2O2 homeodynamics in heart mitochondria.
Collapse
Affiliation(s)
- Pius Abraham Tetteh
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, PE, Canada
| | - Zahra Kalvani
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, PE, Canada
| | - Don Stevens
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, PE, Canada
| | - Ravinder Sappal
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, PE, Canada; Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, New York, USA
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, PE, Canada.
| |
Collapse
|
46
|
Zheng Z, Fang J, Shen Y, Mi C, Xu Z, Zhao J, Chen W, Han R, Lei Q, Zhang H. Copper exposure induces trophoblast cell cuproptosis by up-regulating lnc-HZ11. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 281:116641. [PMID: 38936053 DOI: 10.1016/j.ecoenv.2024.116641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 06/29/2024]
Abstract
Copper pollution has attracted global environmental concern. Widespread Cu pollution results in excessive Cu accumulation in human. Epidemiological studies and animal experiments revealed that Cu exposure might have reproductive toxicity. Cuproptosis is a recently reported Cu-dependent and programmed cell death pattern. However, the mechanism by which copper exposure might cause cell cuproptosis is largely unknown. We chose trophoblast cells as cell model and found that copper exposure causes trophoblast cell cuproptosis. In mechanism, copper exposure up-regulates lnc-HZ11 expression levels, which increases intracellular Cu2+ levels and causes trophoblast cell cuproptosis. Knockdown of lnc-HZ11 efficiently reduces intracellular Cu2+ levels and alleviate trophoblast cell cuproptosis, which could be further alleviated by co-treatment with DC or TEPA. These results discover novel toxicological effects of copper exposure and also provide potential target for protection trophoblast cells from cuproptosis in the presence of excessive copper exposure.
Collapse
Affiliation(s)
- Zhaodian Zheng
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Jing Fang
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yanqiu Shen
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Chenyang Mi
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Zhongyan Xu
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Jingsong Zhao
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Weina Chen
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Ruining Han
- Department of Obstetrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China.
| | - Qiong Lei
- Department of Obstetrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China.
| | - Huidong Zhang
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China.
| |
Collapse
|
47
|
Wang H, Xie M, Zhao Y, Zhang Y. Establishment of a prognostic risk model for prostate cancer based on Gleason grading and cuprotosis related genes. J Cancer Res Clin Oncol 2024; 150:376. [PMID: 39085482 PMCID: PMC11291559 DOI: 10.1007/s00432-024-05899-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION Prostate cancer (PCa) is common in aging males, diagnosed via the Gleason grading system. The study explores the unexamined prognostic value of cuprotosis, a distinct cell death type, alongside Gleason grades in PCa. METHODS We explored Cuprotosis-related genes (CRGs) in prostate cancer (PCa), using NMF on TCGA-PRAD data for patient classification and WGCNA to link genes with Gleason scores and prognosis. A risk model was crafted via LASSO Cox regression. STX3 knockdown in PC-3 cells, analyzed for effects on cell behaviors and tumor growth in mice, highlighted its potential therapeutic impact. RESULTS We identified five genes crucial for a prognostic risk model, with higher risk scores indicating worse prognosis. Survival analysis and ROC curves confirmed the model's predictive accuracy in TCGA-PRAD and GSE70769 datasets. STX3 was a key adverse prognostic factor, with its knockdown significantly reducing mRNA and protein levels, impairing PC-3 cell functions. In vivo, STX3 knockdown in PC-3 cells led to significantly smaller tumors in nude mice, underscoring its potential therapeutic value. CONCLUSION Our prognostic model, using five genes linked to Gleason scores, effectively predicts prostate cancer outcomes, offering a novel treatment strategy angle.
Collapse
Affiliation(s)
- Haicheng Wang
- Department of Urology, Hebei Medical University, Shijiazhuang, China
- Department of Urology, Qinhuangdao First Hospital, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, China
| | - Meiyi Xie
- Department of Urology, Qinhuangdao First Hospital, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, China
| | - Yuming Zhao
- Department of Urology, Qinhuangdao First Hospital, No. 258 Wenhua Road, Haigang District, Qinhuangdao, 066000, China
| | - Yong Zhang
- Department of Urology, Hebei Medical University, Shijiazhuang, China.
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
48
|
Jia D, Liu L, Liu W, Li J, Jiang X, Xin Y. Copper metabolism and its role in diabetic complications: A review. Pharmacol Res 2024; 206:107264. [PMID: 38876443 DOI: 10.1016/j.phrs.2024.107264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/03/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Disturbances in copper (Cu) homeostasis have been observed in diabetes and associated complications. Cu is an essential micronutrient that plays important roles in various fundamental biological processes. For example, diabetic cardiomyopathy is associated with elevated levels of Cu in the serum and tissues. Therefore, targeting Cu may be a novel treatment strategy for diabetic complications. This review provides an overview of physiological Cu metabolism and homeostasis, followed by a discussion of Cu metabolism disorders observed during the occurrence and progression of diabetic complications. Finally, we discuss the recent therapeutic advances in the use of Cu coordination complexes as treatments for diabetic complications and their potential mechanisms of action. This review contributes to a complete understanding of the role of Cu in diabetic complications and demonstrates the broad application prospects of Cu-coordinated compounds as potential therapeutic agents.
Collapse
Affiliation(s)
- Dongkai Jia
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy and Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Science, Jilin University, Changchun 130021, China
| | - Lulu Liu
- Department of Emergency and Critical Medicine, the Second Hospital of Jilin University, Changchun 130012, China
| | - Wei Liu
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Science, Jilin University, Changchun 130021, China
| | - Jinjie Li
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Science, Jilin University, Changchun 130021, China
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy and Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China.
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Science, Jilin University, Changchun 130021, China.
| |
Collapse
|
49
|
Zhao R, Sukocheva O, Tse E, Neganova M, Aleksandrova Y, Zheng Y, Gu H, Zhao D, Madhunapantula SV, Zhu X, Liu J, Fan R. Cuproptosis, the novel type of oxidation-induced cell death in thoracic cancers: can it enhance the success of immunotherapy? Cell Commun Signal 2024; 22:379. [PMID: 39068453 PMCID: PMC11282696 DOI: 10.1186/s12964-024-01743-2] [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: 02/26/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Copper is an important metal micronutrient, required for the balanced growth and normal physiological functions of human organism. Copper-related toxicity and dysbalanced metabolism were associated with the disruption of intracellular respiration and the development of various diseases, including cancer. Notably, copper-induced cell death was defined as cuproptosis which was also observed in malignant cells, representing an attractive anti-cancer instrument. Excess of intracellular copper leads to the aggregation of lipoylation proteins and toxic stress, ultimately resulting in the activation of cell death. Differential expression of cuproptosis-related genes was detected in normal and malignant tissues. Cuproptosis-related genes were also linked to the regulation of oxidative stress, immune cell responses, and composition of tumor microenvironment. Activation of cuproptosis was associated with increased expression of redox-metabolism-regulating genes, such as ferredoxin 1 (FDX1), lipoic acid synthetase (LIAS), lipoyltransferase 1 (LIPT1), dihydrolipoamide dehydrogenase (DLD), drolipoamide S-acetyltransferase (DLAT), pyruvate dehydrogenase E1 subunit alpha 1 (PDHA1), and pyruvate dehydrogenase E1 subunit beta (PDHB)). Accordingly, copper-activated network was suggested as an attractive target in cancer therapy. Mechanisms of cuproptosis and regulation of cuproptosis-related genes in different cancers and tumor microenvironment are discussed in this study. The analysis of current findings indicates that therapeutic regulation of copper signaling, and activation of cuproptosis-related targets may provide an effective tool for the improvement of immunotherapy regimens.
Collapse
Affiliation(s)
- Ruiwen Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Olga Sukocheva
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia.
| | - Edmund Tse
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia
| | - Margarita Neganova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yulia Aleksandrova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yufei Zheng
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Gu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Deyao Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - SabbaRao V Madhunapantula
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Xiaorong Zhu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Junqi Liu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ruitai Fan
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
50
|
Zou X, Liu X, Wang H, Li Z, Zhou C. Characterization of cuproptosis signature in clear cell renal cell carcinoma by single cell and spatial transcriptome analysis. Discov Oncol 2024; 15:300. [PMID: 39044005 PMCID: PMC11266328 DOI: 10.1007/s12672-024-01162-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024] Open
Abstract
Cuproptosis is a novel type to regulate cell death with copper-dependent manner, and has been reported to involve in the occurrence and development of various malignant tumors. However, the association between cuproptosis and the tumor microenvironment (TME) of clear cell renal cell carcinoma (ccRCC) remained unclear. To address this question, we integrated the single cell RNA sequencing (scRNA-seq) datasets of ccRCC across different stages, systematically examined the distinctive expression patterns of cuproptosis-related genes (CRGs) within the TME of ccRCC, and explored the crucial signatures using the spatial transcriptome sequencing (ST-seq) dataset. The cuproptosis activities reduced in cancer tissues along with the ccRCC development, and recovered after therapy. We identified HILPDA+ ccRCC1 subtype, characterized with hypoxia, as cuproptosis susceptible cells associated with a better prognosis. The main co-expression modules of HILPDA+ ccRCC1 subtype highlighted the role in anion transport, response to oxygen species and PD-L1-PD-1 pathway. Furthermore, the immunosuppressive cells might interact with HILPDA+ ccRCC1 subtype via HAVCR2-LGALS9, C3-C3AR1, HLA-A-CD8B and HLA-C-CD8A axises to shape the cuproptosis-related TME landscape. In summary, we anticipate that this study will offer valuable insights and potential strategies of cuproptosis for therapy of ccRCC.
Collapse
Affiliation(s)
- Xiaohong Zou
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Xiaoqing Liu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Huiting Wang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Zhenhua Li
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Chen Zhou
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| |
Collapse
|