1
|
Liu J, Tang W, Chen L, Zhang Q, Liu T, Qin L, Zhang Y, Chen X. Engineered gold nanoparticles for accurate and full-scale tumor treatment via pH-dependent sequential charge-reversal and copper triggered photothermal-chemodynamic-immunotherapy. Biomaterials 2025; 321:123322. [PMID: 40222257 DOI: 10.1016/j.biomaterials.2025.123322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/12/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025]
Abstract
Current anti-tumor strategies majorly rely on the targeted delivery of functional nanomedicines to tumor region, neglecting the importance of effective infiltration of these nanomedicines in whole tumor tissue. This process normally causes the quick endocytosis by the tumor cells at surface layer of tumor tissue, resulting in the restriction of the penetration of these nanomedicines and limited therapeutic region, which would not be able to treat the entire tumor tissue. Herein, we prepared a series of engineered gold nanoparticles (Au-MBP NPs) with step-wise charge reversal in different acid environments that could entirely infiltrate into the whole tumor tissue and then perform tumor-specific photothermal-chemodynamic-immunotherapy to achieve the complete and accurate tumor treatment. These Au-MBP NPs consisted of AuNPs, thiol modified piperidine (SH-PD, charge reversal group), thiol modified benzoyl thiourea (SH-BTU, copper chelator) and 11-mercaptoundecanoic acid (MUA) with different proportions. Once these Au-MBP NPs arrived tumor tissue, the decreasing pH values from shallow to deep region of tumor tissue separately induced the charge reversal of these nanoparticles from negative to positive, allowing them to bind with negatively charged tumor cells at designed area to occupy the whole tumor for further therapy. Following with the internalization by tumor cells, these Au-MBP NPs would selectively capture the excessive Cu2+ to decrease the available copper in tumor cells, resulting in the inhibition of tumor metastasis via the copper metabolism blockade. On one hand, the captured Cu2+ also induced the aggregation of Au-MBP NPs, which in situ generated the photothermal agents in tumor cells for tumor-specific photothermal therapy (PTT). On the other hand, the chelated Cu2+ ions were reduced to Cu+, which catalyzed the high concentration of intracellular H2O2 to produce cytotoxic hydroxyl radical (•OH), exerting tumor-specific chemodynamic therapy (CDT). Furthermore, the immune-associated tumor antigens were also generated during PTT and CDT processes via immunogenic cell death (ICD), which further matured the dendritic cells (DCs) and then activated CD4+ and CD8+ T cells to turn on the immunotherapy, resulting in additional anti-tumor and anti-metastasis effects. Both in vitro and in vivo results indicated that these Au-MBP NPs possessed enormous potential for effectively suppressing primary and metastatic tumors.
Collapse
Affiliation(s)
- Jie Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Wenjuan Tang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China; School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Li Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qianqian Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tao Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Longyu Qin
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
2
|
Singh J, Srikrishna S. Scribble knockdown induced metastasis, identification of its associated novel molecular candidates through proteome studies. Biochem Biophys Res Commun 2025; 769:151999. [PMID: 40367906 DOI: 10.1016/j.bbrc.2025.151999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/07/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Metastasis is the primary cause of cancer associated deaths globally. Loss of function of Scribble, a cell polarity regulator and tumor suppressor gene, is associated with many forms of human cancers but its role in cell proliferation and metastasis remains unknown. We generated metastatic cancer condition in Drosophila using UASRNAi-GAL4 system by knockdown of Scribble in the wing imaginal discs and tracked metastasis events from early to late pupae (0hr-84 h s) using fluorescence microscopy. Here, we report, for the first time, that the knockdown of Scribble alone could lead to the development of primary tumor in the wing imaginal discs, which is capable of establishing metastasis, apparently leading to secondary tumor formation in pupae at early stage, eventually resulting in absolute pupal lethality without organ development. MMP1, a metastasis biomarker, levels were assessed during pre-and post-metastatic phases in pupae using qRT-PCR and Western blot analysis. Further, we analyzed the proteome of Scribble knockdown induced tumor-bearing pupae by 2-D gel electrophoresis followed by MALDI-TOF MS to identify some novel proteins possibly involved in the progression of tumorigenesis and metastasis events. Six differentially expressed proteins, Obp 99b, Fer2LCH,CG13492, Hsp23, Ubiquitin and Colt, were identified in Scrib knockdown pupae and validated their expression using qRT-PCR. Thus, our results suggested that loss of Scrib alone capable of causing metastasis, without the need for cooperative interaction with oncogenic Ras. The newly identified proteins could be important candidates for biomarker/therapeutic target against Scrib associated metastatic cancers.
Collapse
Affiliation(s)
- Jyotsna Singh
- Cancer and Neurobiology Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Saripella Srikrishna
- Cancer and Neurobiology Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
3
|
Bhatnagar K, Raju S, Patki N, Motiani RK, Chaudhary S. Targeting mineral metabolism in cancer: Insights into signaling pathways and therapeutic strategies. Semin Cancer Biol 2025; 112:1-19. [PMID: 40024314 DOI: 10.1016/j.semcancer.2025.02.011] [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: 07/30/2024] [Revised: 01/29/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
Cancer remains the second leading cause of death worldwide, emphasizing the critical need for effective treatment and control strategies. Essential minerals such as copper, iron, zinc, selenium, phosphorous, calcium, and magnesium are integral to various biological processes and significantly influence cancer progression through altered metabolic pathways. For example, dysregulated copper levels promote tumor growth, while cancer cells exhibit an increased dependency on iron for signaling and redox reactions. Zinc influences tumor development through pathways such as Akt-p21. Selenium, primarily through its role in selenoproteins, exhibits anticancer potential but may also contribute to tumor progression. Similarly, dietary phosphate exacerbates tumorigenesis, metastasis, and angiogenesis through signaling pathway activation. Calcium, the most abundant mineral in the body, is tightly regulated within cells, and its dysregulation is a hallmark of various cancers. Magnesium deficiency, on the other hand, promotes cancer progression by fostering inflammation and free radical-induced DNA mutations. Interestingly, magnesium also plays a dual role, with low levels enhancing epithelial-mesenchymal transition (EMT), a critical process in cancer metastasis. This complex interplay of essential minerals underscores their potential as therapeutic targets. Dysregulation of these minerals and their pathways could be exploited to selectively target cancer cells, offering novel therapeutic strategies. This review summarizes current research on the abnormal accumulation or depletion of these microelements in tumor biology, drawing evidence from animal models, cell lines, and clinical samples. We also highlight the potential of these minerals as biomarkers for cancer diagnosis and prognosis, as well as therapeutic approaches involving metal chelators, pharmacological agents, and nanotechnology. By highlighting the intricate roles of these minerals in cancer biology, we aim to inspire further research in this critical yet underexplored area of oncology.
Collapse
Affiliation(s)
- Kartik Bhatnagar
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Plot Nos. 8-11, Tech Zone 2, Greater Noida, Uttar Pradesh 201310, India.
| | - Sharon Raju
- Laboratory of Calciomics and Systemic Pathophysiology, Regional Centre for Biotechnology (RCB), Faridabad-Gurugram Expressway, Faridabad, Haryana 121001, India.
| | - Ninad Patki
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Plot Nos. 8-11, Tech Zone 2, Greater Noida, Uttar Pradesh 201310, India.
| | - Rajender K Motiani
- Laboratory of Calciomics and Systemic Pathophysiology, Regional Centre for Biotechnology (RCB), Faridabad-Gurugram Expressway, Faridabad, Haryana 121001, India.
| | - Sarika Chaudhary
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Plot Nos. 8-11, Tech Zone 2, Greater Noida, Uttar Pradesh 201310, India.
| |
Collapse
|
4
|
Mao L, Lu J, Wen X, Song Z, Sun C, Zhao Y, Huang F, Chen S, Jiang D, Che W, Zhong C, Yu C, Li K, Lu X, Shi J. Cuproptosis: mechanisms and nanotherapeutic strategies in cancer and beyond. Chem Soc Rev 2025. [PMID: 40433941 DOI: 10.1039/d5cs00083a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
Cuproptosis, a novel form of copper (Cu)-dependent programmed cell death, is induced by directly binding Cu species to lipoylated components of the tricarboxylic acid (TCA) cycle. Since its discovery in 2022, cuproptosis has been closely linked to the field of materials science, offering a biological basis and bright prospects for the use of Cu-based nanomaterials in various disease treatments. Owing to the unique physicochemical properties of nanomaterials, Cu delivery nanosystems can specifically increase Cu levels at disease sites, inducing cuproptosis to achieve disease treatment while minimizing the undesirable release of Cu in normal tissues. This innovative nanomaterial-mediated cuproptosis, termed as "nanocuproptosis", positions at the intersection of chemistry, materials science, pharmaceutical science, and clinical medicine. This review aims to comprehensively summarize and discuss recent advancements in cuproptosis across various diseases, with a particular focus on cancer. It delves into the biochemical basis of nanomaterial-mediated cuproptosis, the rational design for cuproptosis inducers, strategies for enhancing therapeutic specificity, and cuproptosis-centric synergistic cancer therapeutics. Beyond oncology, this review also explores the expanded applications of cuproptosis, such as antibacterial, wound healing, and bone tissue engineering, highlighting its great potential to open innovative therapeutic strategies. Furthermore, the clinical potential of cuproptosis is assessed from basic, preclinical to clinical research. Finally, this review addresses current challenges, proposes potential solutions, and discusses the future prospects of this burgeoning field, highlighting cuproptosis nanomedicine as a highly promising alternative to current clinical therapeutics.
Collapse
Affiliation(s)
- Lijie Mao
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Ji Lu
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xinyu Wen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Zhiyi Song
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Cai Sun
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yuanru Zhao
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Fang Huang
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Si Chen
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Dongyang Jiang
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Cheng Zhong
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Ke Li
- School of Materials Science and Engineering, Hainan University, Haikou 570228, China.
| | - Xiangyu Lu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Jianlin Shi
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| |
Collapse
|
5
|
Lee J, Kim T. Current Status and Future Perspectives of Nuclear Medicine in Prostate Cancer from Imaging to Therapy: A Comprehensive Review. Biomedicines 2025; 13:1132. [PMID: 40426959 DOI: 10.3390/biomedicines13051132] [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: 03/28/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Nuclear medicine has emerged as a critical modality in the diagnostic and therapeutic management of urological malignancies, particularly prostate cancer. Advances in single-photon emission computed tomography/computed tomography (CT) and positron emission tomography/CT (PET/CT) have enhanced tumor assessment across staging, treatment response, and recurrence settings. Molecular imaging, which offers insights beyond traditional anatomical imaging, is increasingly integral in specific clinical scenarios. Theranostic nuclear medicine, which combines diagnostic imaging with targeted therapy, has become a well-established treatment option, particularly for patients with metastatic castration-resistant prostate cancer (mCRPC). The development of the prostate-specific membrane antigen (PSMA) radioligands has revolutionized clinical management by enabling precise disease staging and delivering effective radioligand therapy (RLT). Ongoing research aims to refine the role of PSMA PET imaging in staging and treatment monitoring, while optimizing PSMA-targeted RLT for broader clinical use. Given that prostate cancer remains highly prevalent, the anticipated increase in the demand for RLT presents both challenges and opportunities for nuclear medicine services globally. Theranostic approaches exemplify personalized medicine by enabling the tailoring of treatments to individual tumor biology, thereby improving survival outcomes and maintaining patients' quality of life with minimal toxicity. Although the current focus is on advanced disease, future research holds promise for expanding these strategies to earlier stages, potentially enhancing curative prospects. This evolving field not only signifies a paradigm shift in the care of prostate cancer patients but also underscores the growing importance of nuclear medicine in delivering precision oncology.
Collapse
Affiliation(s)
- Joohee Lee
- CHA Ilsan Medical Center, Department of Nuclear Medicine, CHA University College of Medicine, Ilsan 10414, Gyeonggi-do, Republic of Korea
| | - Taejin Kim
- CHA Ilsan Medical Center, Department of Urology, CHA University College of Medicine, Ilsan 10414, Gyeonggi-do, Republic of Korea
| |
Collapse
|
6
|
Cheng X, Hemmati S, Pirhayati M, Zangeneh MM, Veisi H. Decoration of copper nanoparticles (Cu 2O NPs) over chitosan-guar gum: Its application in the Sonogashira cross-coupling reactions and treatment of human lung adenocarcinoma. Int J Biol Macromol 2025; 305:141122. [PMID: 39965696 DOI: 10.1016/j.ijbiomac.2025.141122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/02/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
This study outlines the sustainable synthesis of hybrid biopolymer hydrogels supported with octahedral Cu2O nanoparticles (NPs), alongside their biological assessments and characterizations. A composite hydrogel made of chitosan and guar gum (CS-GG) was employed as a template for the environmentally friendly synthesis of nanoparticles. Leveraging their electron-rich functional groups, the biopolymers acted as stabilizing agents for the Cu2O NPs and as green reductants, facilitating the reduction of copper ions. The material's physicochemical properties were thoroughly examined using advanced techniques, such as X-ray diffraction (XRD), Field-Emission Scanning Electron Microscopes (FE-SEM), Eneregy Dispersive X-ray Electron Spectroscopy (EDX), Fourier Transformed Infrared Spectroscopy (FT-IR), Transmission Electron Microscopy (TEM) and ICP-OES. The resulting CS-GG/Cu2O NPs nanocomposite was investigated as a reusable heterogeneous nanocatalyst, demonstrating its efficiency in the phosphine-free, palladium-free, and ligand-free synthesis of various stilbene derivatives with high yields through the Sonogashira coupling reaction. The catalyst showed no significant reduction in activity after being reused seven times consecutively. The cytotoxic effects of the CS-GG/Cu2O NPs nanocomposite on NCI-H661 lung cancer cells and normal cells (HUVEC) were assessed over 48 h using MTT assay. The cancer cell's viability decreased after exposure to the CS-GG/Cu2O NPs, with an IC50 value of 82 μg/mL. The CS-GG/Cu2O NPs nanocomposite controls the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) system, which in turn impacts apoptosis and cell proliferation in NCI-H661 cells, according to a detailed examination of the mTOR pathway. The pathway could act a role in the cell cycle inhibition and apoptosis induced by the CS-GG/Cu2O NPs nanocomposite. The CS-GG/Cu2O NPs nanocomposite could be a useful natural anti-cancer agent for the treatment of lung cancer.
Collapse
Affiliation(s)
- Xiongtao Cheng
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Saba Hemmati
- Department of Chemistry, Payame Noor University, Tehran, Iran
| | - Mozhgan Pirhayati
- Department of Applied Chemistry, Faculty of Science, Malayer University, Malayer, Iran.
| | - Mohamad Mehdi Zangeneh
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Hojat Veisi
- Department of Chemistry, Payame Noor University, Tehran, Iran.
| |
Collapse
|
7
|
Sun Q, Liu H, Yang Y, Yao S, Liu Z, Guo Z. A Self-Immobilizing Photoacoustic Probe for Ratiometric In Vivo Imaging of Cu(II) in Tumors. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:260-266. [PMID: 40313533 PMCID: PMC12041948 DOI: 10.1021/cbmi.4c00115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 05/03/2025]
Abstract
Cu(II) ions play a critical role in tumor growth and metastasis, making in vivo high-resolution imaging of Cu(II) crucial for understanding its role in tumor pathophysiology. However, designing suitable molecular probes for this purpose remains challenging. Herein, we report the development of a photoacoustic probe for specific in vivo imaging of Cu(II) in tumors. This probe utilizes β-galactoside as a targeting group and incorporates a unique self-immobilization strategy. Upon β-galactosidase-mediated cleavage, the probe generates a reactive quinone methide intermediate that covalently binds to intracellular proteins, enabling selective tumor accumulation. The probe exhibits a ratiometric photoacoustic response to Cu(II) with high selectivity over that of other biological species. In vitro and in vivo studies demonstrated the efficacy of the probe for Cu(II) imaging in tumors. This research provides valuable insights into the role of Cu(II) in tumorigenesis and may facilitate the development of diagnostic and therapeutic approaches for cancer.
Collapse
Affiliation(s)
- Qian Sun
- Chemistry
and Biomedicine Innovation Center (ChemBIC), State Key Laboratory
of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023 China
| | - Hang Liu
- College
of Materials Science and Engineering, College of Science, Co-Innovation
Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China
| | - Ying Yang
- Chemistry
and Biomedicine Innovation Center (ChemBIC), State Key Laboratory
of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023 China
| | - Shankun Yao
- Chemistry
and Biomedicine Innovation Center (ChemBIC), State Key Laboratory
of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023 China
| | - Zhipeng Liu
- College
of Materials Science and Engineering, College of Science, Co-Innovation
Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China
| | - Zijian Guo
- Chemistry
and Biomedicine Innovation Center (ChemBIC), State Key Laboratory
of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023 China
| |
Collapse
|
8
|
Wu S, Xia X, Zhou R, Zhao H. Hydrogel-enabled ROS-GSH modulation for sustained copper-mediated chemodynamic therapy of oral squamous cell carcinoma. J Control Release 2025; 383:113772. [PMID: 40280240 DOI: 10.1016/j.jconrel.2025.113772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Copper ion (Cu2+) has been revealed to be involved in the occurrence and development of oral squamous cell carcinoma (OSCC), making copper-mediated chemodynamic therapy (Cu-CDT) a promising treatment strategy for OSCC by elevating Cu2+ levels to generating a large amount of reactive oxygen species (ROS). However, the excessive reduced glutathione (GSH) in the tumor microenvironment can scavenge the ROS generated by Cu-CDT. While the directional co-delivery of Cu2+ and GSH-depleting agents shows promise for Cu-CDT in OSCC therapy, their rapid metabolism and the superficial nature of OSCC lesions necessitate tailored drug formulations to ensure effective bioavailability. To counteract this challenge, this work proposed a practical hydrogel-supported ROS-GSH regulation strategy, which involves the on-demand design of a copper ion-crosslinked guanosine-based hydrogel (GCD) containing dimethyl fumarate (DMF, which conjugates with GSH for consumption). It can directionally and sustainably co-deliver Cu2+ and DMF to OSCC lesions under mildly acidic pH conditions, thereby enhancing Cu-CDT efficiency through improved Cu2+ utilization and DMF-driven GSH depletion. As anticipated, the strategy sustains the generation of hydroxyl radicals, effectively inducing apoptosis and suppressing cell proliferation in CAL-27 cells, which consequently inhibits the growth of OSCC tumors. Therefore, this work highlights the GCD hydrogel's great potential as a promising Cu-CDT therapeutic platform.
Collapse
Affiliation(s)
- Shihong Wu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xin Xia
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ronghui Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Hang Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Gui W, Wang WX. Intestinal Cu(II)/(I) Redox State Transformation Causes Cu(I) Overflow and Toxicity of the Gut and Liver in Zebrafish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:7495-7505. [PMID: 40223213 DOI: 10.1021/acs.est.4c14690] [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: 04/15/2025]
Abstract
Copper (Cu) has long been a concern for human health. While previous studies have explored the toxic effects of Cu, no study is available on the relationship between the Cu redox state transformation and biotoxicity in higher organisms. In this study, we explored the gut and liver toxicity caused by the overflow of Cu(I) at low doses of Cu exposure. Here, we first elucidated the digestive and metabolic systems as the main toxic target sites by a systematic epidemiological analysis. Then, ICP-MS analysis verified that the gut and liver were the top two Cu-high-accumulated organs in zebrafish exposed to 10 and 100 μg/L waterborne Cu for 72 h. In-situ Cu(I) and Cu(II) imaging techniques demonstrated that exogenous Cu(II) was converted to Cu(I) in the zebrafish gut. Furthermore, transcriptomic sequencing revealed that the high overflow of Cu(I) induced gut toxicity by cell cycle arrest in the G phase. However, the substantial accumulation of Cu(I) disrupted the metabolism of energy source nutrients and energy supply, leading to hepatic toxicity. This study provides new insights into the toxic mechanism based on Cu redox state and emphasizes the health risks associated with Cu exposure in the digestive and metabolic systems.
Collapse
Affiliation(s)
- Wanying Gui
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong(852)95772465, China
- Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Wen-Xiong Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong(852)95772465, China
- Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
10
|
Li Y, Han L, Hu H. Research progress on cuproptosis and copper related anti-tumor therapy. Discov Oncol 2025; 16:584. [PMID: 40257639 PMCID: PMC12011693 DOI: 10.1007/s12672-025-02335-3] [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: 04/06/2024] [Accepted: 04/08/2025] [Indexed: 04/22/2025] Open
Abstract
Copper is a trace element which is essential for biological organisms, and its homeostatic balance is important for living organisms to maintain the normal function. When the copper homeostasis is disordered, the cellular function and structure will be disrupted. Excess copper cause oxidative stress and DNA damage in cells, thereby inducing regulated cell death such as apoptosis and necroptosis. Excess copper in mitochondria can bind to lipoylated proteins in the tricarboxylic acid (TCA) cycle and cause them to aggregate, resulting in proteotoxic stress and eliciting a novel cell death modality: cuproptosis. Cancer cells have a greater demand for copper compared to normal tissue, and high levels of copper ions are closely associated with tumour proliferation and metastasis. The anti-tumor mechanisms of copper include the production of oxidative stress, inhibition of the ubiquitin-proteasome system, suppression of angiogenesis, and induction of copper-dependent cell death. Targeting copper is one of the current directions in oncology research, including the use of copper ion carriers to increase intracellular copper levels to induce oxidative stress and cuproptosis, as well as the use of copper ion chelators to reduce copper bioavailability. However, copper complexes have certain toxicity, so their biosafety needs to be improved. Emerging nanotechnology is expected to solve this problem by utilizing copper-based nanomaterials (Cu-based NMs) to deliver copper ions and a variety of drugs with different functions, thereby improving the anti-tumor efficacy and reducing the side effects. Therefore, a thorough understanding of copper metabolic processes and the mechanism of cuproptosis will greatly benefit anti-tumor therapy. This review summarizes the processes of copper metabolism and the mechanism of cuproptosis. In addition, we discuss the current anti-tumor paradigms related to copper, we also discuss current nanotherapeutic approaches to copper mortality and provide prospective insights into the future copper-mediated cancer therapy.
Collapse
Affiliation(s)
- Yichen Li
- School of Medicine, Southeast University, No. 87, Dingjiaqiao, Hunan Road, Gulou District, Nanjing, 210009, China
| | - Lifei Han
- Breast Disease Diagnosis and Treatment Center, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, China
| | - Haolin Hu
- Breast Disease Diagnosis and Treatment Center, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, China.
| |
Collapse
|
11
|
Wang Z, Zeng Y, Gao S, Chen Z, Chen C, Wang Y. Naphthazarin Mounted on the Manganese Carbonate Nanocube Induced Enrichment of Endogenous Copper and Fenton-like Reaction for Enhanced Chemodynamic Therapy. ACS APPLIED BIO MATERIALS 2025; 8:3309-3320. [PMID: 40129267 DOI: 10.1021/acsabm.5c00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Chemodynamic therapy (CDT), which utilizes transition metal ions to catalyze Fenton-like reactions for the eradication of tumor cells, has attracted substantial attention in the field of nanocatalysis. However, the therapeutic efficacy of CDT as a monotherapy is often limited by an insufficient level of hydrogen peroxide (H2O2) and the overexpressed glutathione (GSH) within tumor cells. Because of the high copper content in tumor tissues, a copper ionophore was strategically employed to enhance the intracellular accumulation of copper, thereby potentiating the CDT effect. Additionally, bovine serum albumin (BSA) was used to modify the copper ionophore, naphthazarin (Nap), to promote its targeting efficacy for tumor cells and to ensure its biosafety. The BSA-coated Nap nanoparticles, which could recruit Cu2+ in situ, were further deposited onto the surface of a manganese carbonate nanocube (Nap-BM NPs). The synergistic action of copper and manganese ions accelerated the decomposition of H2O2 into hydroxyl radicals (•OH) and consumed intracellular GSH, leading to cellular mortality via mitochondrial pathways. With low cytotoxicity and good biocompatibility in normal cells, the developed Nap-BM NPs significantly enhanced therapeutic outcomes by leveraging multiple Fenton-like reaction mechanisms to augment CDT, offering promising potential for clinical applications and contributing valuable insights into the field.
Collapse
Affiliation(s)
- Zhichao Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Yuan Zeng
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Susu Gao
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ziwei Chen
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunying Chen
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaling Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
12
|
Zhang B, Zheng D, Zhu S, Zhang X, Wang Q, Lin Z, Zheng Z, Zhou S, Chen Z, Zheng S, Lan E, Cui L, Ying H, Zhang Y, Lin X, Zhuang Q, Qian H, Hu X, Zhuang Y, Zhang Q, Jin Z, Jiang S, Ma Y. Leveraging a disulfidptosis-based signature to characterize heterogeneity and optimize treatment in multiple myeloma. Front Immunol 2025; 16:1559317. [PMID: 40308607 PMCID: PMC12041008 DOI: 10.3389/fimmu.2025.1559317] [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: 01/12/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Background Disulfidptosis is an emerging type of programmed cell death related to ROS accumulation and aberrant disulfide bond formation. Multiple myeloma (MM) is the second most prevalent hematologic malignancy characterized by a high synthesis rate of disulfide bond-rich proteins and chronic oxidative stress. However, the relationship between disulfidptosis and MM is still unclear. Methods Using the non-negative matrix factorization and lasso algorithm, we constructed the disulfidptosis-associated subtypes and the prognostic model on the GEO dataset. We further explored genetic mutation mapping, protein-protein interactions, functional enrichment, drug sensitivity, drug prediction, and immune infiltration analysis among subtypes and risk subgroups. To improve the clinical benefits, we combined risk scores and clinical metrics to build a nomogram. Finally, in vitro experiments examined the expression patterns of disulfidptosis-related genes (DRGs) in MM. Results By cluster analysis, we obtained three subtypes with C2 having a worse prognosis than C3. Consistently, C2 exhibited significantly lower sensitivity to doxorubicin and lenalidomide, as well as a higher propensity for T-cell depletion and a non-responsive state to immunotherapy. Similarly, in the subsequent prognostic model, the high-scoring group had a worse prognosis and a higher probability of T-cell dysfunction, immunotherapy resistance, and cancer cell self-renewal. DRGs and risk genes were widely mutated in cancers. Subtypes and risk subgroups differed in ROS metabolism and the p53 signaling pathway. We further identified eight genes differentially expressed in risk subgroups as drug targets against MM. Then 27 drugs targeting the high-risk group were predicted. Based on the DRGs and risk genes, we constructed the miRNA and TF regulatory networks. The nomogram of combined ISS, age, and risk score showed good predictive performance. qRT-PCR of cell lines and clinical specimens provided further support for prognostic modeling. Conclusion Our research reveals the prognostic value of disulfidptosis in MM and provides new perspectives for identifying heterogeneity and therapeutic targets.
Collapse
Affiliation(s)
- Bingxin Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dong Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuxia Zhu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyi Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Quanqiang Wang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhili Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziwei Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shujuan Zhou
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zixing Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sisi Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Enqing Lan
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Luning Cui
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hansen Ying
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yu Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuanru Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiang Zhuang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Honglan Qian
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xudong Hu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Zhuang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qianying Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouxiang Jin
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Songfu Jiang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongyong Ma
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang, Wenzhou, Zhejiang, China
- Zhejiang Engineering Research Center for Hospital Emergency and Process Digitization, Wenzhou, Zhejiang, China
| |
Collapse
|
13
|
Fernández-González M, Armisen R, Fernández MI. Predator-Prey Model for Simulating the Genetic Carcinogenicity of Aggressive Toxicant-Related Cancer. J Appl Toxicol 2025. [PMID: 40234059 DOI: 10.1002/jat.4790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 04/17/2025]
Abstract
The mechanism of how toxicant exposure leads to aggressive tumors remains unresolved. A genetic-based hypothesis predicts that under stress, the transcription of growth-related genes will be inhibited by the activation of mitogenic pathways, redirecting energy toward stress response and increasing survival. This hypothesis fails to explain why epidemiological data suggest that growth and stress response are activated, as patients exposed to toxicants exhibit more aggressive growth than nonexposed individuals. This co-occurrence requires increased energy availability to prevent the activation of mitogenic pathways, as seen in the Warburg effect. We hypothesize that if pollutant effects cease, it might drive aggressive cancer, as excess energy that is no longer used for stress response can fuel rapid growth. We model this allocation between growth and stress response as a trophic competition using the Lotka-Volterra equations and using as input RNA-Seq data from growth- and stress-related genes obtained from cancer cells exposed to copper, cadmium, and carboplatin. Our findings suggest that the energy allocation to growth and its rate of allocation is higher in exposed than nonexposed tumors and results in overgrowth in unexposed cells. This study helps to understand how certain scenarios, such as partial or total cessation of exposure, in toxicant-related cancer can drive cancer aggressiveness.
Collapse
Affiliation(s)
- Mauricio Fernández-González
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Ricardo Armisen
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Mario I Fernández
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
- Departamento de Urología, Clinica Alemana, Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
14
|
Xiang Z, Mei H, Wang H, Yao X, Rao J, Zhang W, Xu A, Lu L. Cuproptosis and its potential role in musculoskeletal disease. Front Cell Dev Biol 2025; 13:1570131. [PMID: 40292330 PMCID: PMC12022686 DOI: 10.3389/fcell.2025.1570131] [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: 02/03/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Cuproptosis, a recently identified form of copper-dependent cell death, arises from intracellular copper dyshomeostasis. As an essential trace element, copper plays a critical role in bioenergetic metabolism, redox regulation, and synaptic transmission. However, excessive copper exerts cytotoxic effects through multiple pathways, including increased reactive oxygen species (ROS) production, apoptotic cascade activation, necrotic membrane rupture, inflammatory responses, and mitochondrial dysfunction. Distinct from other cell death mechanisms, cuproptosis is characterized by copper ion binding to acetylated mitochondrial respiratory chain proteins, leading to pathogenic protein aggregation, iron-sulfur cluster depletion, and cellular collapse. Emerging evidence underscores aberrant copper accumulation and resultant proteotoxic stress as pivotal contributors to the pathogenesis of multiple musculoskeletal pathologies, including osteoporosis, osteoarthritis, sarcopenia, osteosarcoma, intervertebral disc degeneration, spinal cord injury, and biofilm-associated orthopedic infections. Understanding the spatiotemporal regulation of cuproptosis may provide novel opportunities for advancing diagnostic and therapeutic approaches in orthopedic medicine. This review synthesizes current insights into the molecular mechanisms of cuproptosis, its pathogenic role in musculoskeletal diseases, and the potential for biomarker-driven therapeutic interventions.
Collapse
Affiliation(s)
- Ziyang Xiang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huiling Mei
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Honglin Wang
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyue Yao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ji Rao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wentao Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Aoshuang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Lu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Yi F, Tao S, Wu H. Bilirubin metabolism in relation to cancer. Front Oncol 2025; 15:1570288. [PMID: 40291905 PMCID: PMC12021636 DOI: 10.3389/fonc.2025.1570288] [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: 02/03/2025] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
Bilirubin, a metabolite of hemoglobin, was long thought to be a harmful waste product, but recent studies have found it to have antioxidant and anti-tumor effects. With the extensive research on the mechanism of malignant tumor development, the antioxidant effect of bilirubin is increasingly becoming a hotspot in anti-cancer research. At present, there are two main views on the relationship between bilirubin and cancer, namely, its pro-cancer and anti-cancer effects, and in recent years, studies on the relationship between bilirubin and cancer have not been systematically summarized, which is not conducive to the further investigation of the role of bilirubin on cancer. To understand the multifaceted role of bilirubin in tumorigenesis as well as to develop more effective and affordable antitumor therapies, this review provides an overview of the effects of bilirubin on tumors in terms of oxidative, inflammatory, and cellular signaling pathways, as well as the resulting therapeutic ideas and approaches.
Collapse
Affiliation(s)
- Fengyun Yi
- Department of Traditional Chinese Medicine, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Siyu Tao
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongze Wu
- Department of Traditional Chinese Medicine, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
| |
Collapse
|
16
|
Guan Z, Wang H, Tian M. A Cuproptosis-Related gene Signature as a Prognostic Biomarker in Thyroid Cancer Based on Transcriptomics. Biochem Genet 2025; 63:1584-1604. [PMID: 38594571 DOI: 10.1007/s10528-024-10767-9] [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: 06/09/2023] [Accepted: 02/28/2024] [Indexed: 04/11/2024]
Abstract
Thyroid cancer (THCA) is the most prevalent endocrine tumor, and its incidence continues to increase every year. However, the processes underlying the aggressive progression of thyroid cancer are unknown. We concentrated on the prognostic and biological importance of thyroid cancer cuproptosis-related genes in this investigation. Genomic and clinical data were obtained from the UCSC XENA website, and cuproptosis-related genes were obtained from the FerrDb website. We performed differential expression analysis and Cox regression analysis to identify possible predictive targets associated with thyroid cancer prognosis. To assess the role of CDKN2A in thyroid cancer and the ability to predict prognosis on the basis of the CDKN2A expression level, we performed immunohistochemical staining, survival analysis, immunological analysis, functional analysis, and clinical analysis with respect to CDKN2A gene expression. CDKN2A expression levels were found to be inversely correlated with thyroid cancer prognosis. Higher levels of CDKN2A expression were associated with higher T, N, and clinicopathological stage and more residual tumor cells. Through univariate and multivariate Cox regression analyses, the CDKN2A expression level was shown to be linked with thyroid cancer patients' overall survival (OS). Moreover, we discovered that CDKN2A expression was linked to a dysfunctional tumor immune microenvironment. The study shows that CDKN2A, a cuproptosis-related gene, can be used as a prognostic marker for thyroid cancer.
Collapse
Affiliation(s)
- Zirui Guan
- The Second Hospital of Jilin University, Changchun City, 130022, Jilin Province, People's Republic of China
| | - Hongyong Wang
- The Second Hospital of Jilin University, Changchun City, 130022, Jilin Province, People's Republic of China.
| | - Mingyan Tian
- The Second Hospital of Jilin University, Changchun City, 130022, Jilin Province, People's Republic of China
| |
Collapse
|
17
|
Wu J, Wu W, Qin J, Chen Z, Zhong R, Guo P, Fan S. Exploring the impact of cuproptosis on prostate cancer prognosis via RNA methylation regulation based on single cell and bulk RNA sequencing data. Front Pharmacol 2025; 16:1573611. [PMID: 40235543 PMCID: PMC11996820 DOI: 10.3389/fphar.2025.1573611] [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: 02/09/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Background Cuproptosis, along with RNA methylation regulators, has recently come to the fore as innovative mechanisms governing cell death, exerting profound impact on the onset and progression of multiple cancers. Nonetheless, the prognostic implications and underlying regulatory mechanisms of them associated with prostate cancer (PCa) remain to be thoroughly investigated. Methods Genomic and clinical data for PCa from The Cancer Genome Atlas datasets were analyzed to identify a prognostic model through univariate and Least Absolute Shrinkage and Selection Operator Cox regression analyses that were validated utilizing external datasets. We used receiver operating characteristic curves and C-index to evaluate the accuracy of our prognostic model. In conjunction with this, we conducted single-cell RNA sequencing (scRNA-seq) analyses to investigate underlying mechanisms and evaluate the degree of immune infiltration, as well as to assess patients' responses to diverse chemotherapy agents. Especially, qPCR assay was utilized to unveil the expression of signature genes in PCa. Results We meticulously selected six Cuproptosis-Associated RNA Methylation Regulators (CARMRs) to establish a risk prognosis model, which was further verified to obtain enhanced predictive capacity in external validation cohorts. Insights from immune infiltration and scRNA-seq analyses have elucidated the immune characteristics of PCa, and highlighted the immunosuppressive role of regulatory T cells on immune response. Additionally, drug susceptibility analysis demonstrated that patients with PCa in the low-risk category derived better benefit from bicalutamide treatment, whereas those in the high-risk group exhibited a favor response to adriamycin and docetaxel treatments. The qPCR and immunohistochemistry (IHC) staining assays also reveal the a dramatically altered expression pattern of TRDMT1 and ALYREF in PCa tissues. Conclusion In general, we established a model involving CARMRs that can better predict the risk of recurrence of PCa and have identified the possible mechanisms affecting PCa progression, thereby promoting further research in this field.
Collapse
Affiliation(s)
- Junchao Wu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| | - Wentian Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiaxuan Qin
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| | - Ziqi Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| | - Rongfang Zhong
- Department of Urology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Peng Guo
- Department of Urology, The Affiliated Jiangyin Hospital of Nantong University, Wuxi, China
| | - Song Fan
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| |
Collapse
|
18
|
Valand R, Pandey N, Bellare J, Sivaiah A. A water-soluble glucose-appended quinoline-benzothiazole conjugate as a selective and sensitive receptor for Cu + ions in aqueous media and intracellular bio-imaging in live cells. Analyst 2025; 150:1176-1186. [PMID: 39963841 DOI: 10.1039/d5an00066a] [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: 03/12/2025]
Abstract
A water-soluble and biologically benign glucose-appended quinoline-benzothiazole conjugate (L) has been synthesized and characterized using various spectroscopy techniques. The recognition properties of L show selective recognition of Cu+ ions among other biological metal ions including Cu2+ ions in PBS buffer at pH 7.4. L exhibited a switch-on fluorescence enhancement upon the addition of Cu+ with a detection limit of 1.48 × 10-8 M in an aqueous medium. Job's plot confirmed the 1 : 1 binding ratio observed between probe L and Cu+ ions with an association constant (Ka) of 1.46 × 105 M-1. The proposed complex binding mechanism was supported by UV-Vis, fluorescence and ESI-MS. The coordination features of the {L + Cu+} complex were delineated using DFT computational calculations. Furthermore, L was successfully applied for intracellular fluorescence bio-imaging of Cu+ ions in L929 living cells, suggesting that it holds significant potential for Cu+ ion bio-imaging and disease diagnosis.
Collapse
Affiliation(s)
- Ravinkumar Valand
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat-395007, Gujarat, India.
| | - Nidhi Pandey
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai-400076, Mumbai, India
| | - Jayesh Bellare
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai-400076, Mumbai, India
| | - Areti Sivaiah
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat-395007, Gujarat, India.
| |
Collapse
|
19
|
Wang J, Yang D, Yu HF, Jin J, Nie Y, Zhang S, Ren W, Ge Z, Zhang Z, Ma X, Dai S, Sui G, Teng CB. Copper is essential for cyclin B1-mediated CDK1 activation. Nat Commun 2025; 16:2288. [PMID: 40055333 PMCID: PMC11889272 DOI: 10.1038/s41467-025-57538-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
Cyclin-dependent kinase 1 (CDK1) is the pivotal kinase responsible for initiating cell division. Its activation is dependent on binding to regulatory cyclins, such as CCNB1. Our research demonstrates that copper binding to both CDK1 and CCNB1 is essential for activating CDK1 in cells. Mutations in the copper-binding amino acids of either CDK1 or CCNB1 do not disrupt their interaction but are unable to activate CDK1. We also reveal that CCNB1 facilitates the transfer of copper from ATOX1 to CDK1, consequently activating its kinase function. Disruption of copper transfer through the ATOX1-CCNB1-CDK1 pathway can impede CDK1 activation and halt cell cycle progression. In summary, our findings elucidate a mechanism through which copper promotes CDK1 activation and the G2/M transition in the cell cycle. These results could provide insight into the acquisition of proliferative properties associated with increased copper levels in cancer and offer targets for cancer therapy.
Collapse
Affiliation(s)
- Jiaru Wang
- Laboratory of Cell Biology, College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Dian Yang
- Laboratory of Cell Biology, College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Hai-Fan Yu
- Laboratory of Cell Biology, College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Jing Jin
- Laboratory of Cell Biology, College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Yuzhe Nie
- Laboratory of Cell Biology, College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Sihua Zhang
- Laboratory of Cell Biology, College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Weiwei Ren
- Development Center of Plant Germplasm Resources, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Zihan Ge
- Laboratory of Cell Biology, College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Zhuo Zhang
- Laboratory of Cell Biology, College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Xinghong Ma
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Shaojun Dai
- Development Center of Plant Germplasm Resources, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Guangchao Sui
- Laboratory of Cell Biology, College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Chun-Bo Teng
- Laboratory of Cell Biology, College of Life Science, Northeast Forestry University, Harbin, 150040, China.
| |
Collapse
|
20
|
Frølich A, Dove RE, Friberg M, Behndig AF, Sandström T, Blomberg A, Mudway IS. Respiratory tract lining fluid copper content contributes to pulmonary oxidative stress in patients with systemic sclerosis. Wellcome Open Res 2025; 9:139. [PMID: 40115047 PMCID: PMC11923536 DOI: 10.12688/wellcomeopenres.20080.2] [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] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Background Systemic sclerosis (SSc) is an autoimmune disease characterized by fibrosis of the skin and internal organs, mostly affecting young and middle-aged women. Significant questions remain as to its pathogenesis, especially the triggers for the associated interstitial lung disease (SSc-ILD). We examined the extent to which SSc and SSc-ILD were related to oxidative stress and altered metal homeostasis at the air-lung interface. Methods In this case-control study, we recruited 20 SSc patients, of which 11 had SSc-ILD. Eighteen healthy individuals were recruited as age-matched healthy controls, for a total of 38 study participants. Low molecular weight antioxidants (ascorbate, urate and glutathione), metal transport and chelation proteins (transferrin and ferritin) and metals (Fe and Cu) concentrations, including a measure of the catalytically active metal pool, were determined in respiratory tract lining fluid (RTLF) collected by bronchoalveolar lavage from the SSc group and compared with healthy controls. Results In the SSc group, 14 individuals were of female sex (70%) and the median age was 57 years (range 35-75). We observed evidence of oxidative stress in the RTLFs of SSc patients, characterised by increased concentrations of glutathione disulphide (GSSG, P<0.01), dehydroascorbate (DHA, P<0.05) and urate (P<0.01). This was associated with elevated RTLF Fe (P=0.07) and Cu (P<0.001), and evidence of a catalytic metal pool, demonstrated by an enhanced rate of ascorbate oxidation in the recovered lavage fluid (p<0.01). Cu concentrations were significantly associated with the ascorbate depletion rate (r=0.76, P<0.001), and GSSG (r=0.38, P<0.05) and protein carbonyl (r=0.44, P<0.01) concentrations. Whilst these markers were all increased in SSc patients, we found no evidence for an association with SSc-ILD. Conclusions These data confirm the presence of oxidative stress in the airways of SSc patients and, for the first time, suggest that an underlying defect in metal homeostasis at the air-lung interface may play a role in disease progression.
Collapse
Affiliation(s)
- Andreas Frølich
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Rosamund E Dove
- Wolfson Institute of Population Health, Queen Mary University of London, London, England, UK
| | - Maria Friberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Annelie F Behndig
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Thomas Sandström
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Anders Blomberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Ian S Mudway
- MRC Centre for Environment and Health, Environmental Research Group, Imperial College London, London, England, UK
| |
Collapse
|
21
|
Pellei M, Santini C, Caviglia M, Del Gobbo J, Battocchio C, Meneghini C, Amatori S, Donati C, Zampieri E, Gandin V, Marzano C. Anticancer potential of copper(i) complexes based on isopropyl ester derivatives of bis(pyrazol-1-yl)acetate ligands. RSC Med Chem 2025; 16:849-861. [PMID: 39618961 PMCID: PMC11605304 DOI: 10.1039/d4md00610k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/07/2024] [Indexed: 02/21/2025] Open
Abstract
In this paper, the isopropyl ester derivatives LOiPr and L2OiPr of bis(pyrazol-1-yl)acetic acid and bis(3,5-dimethyl-pyrazol-1-yl)acetic acid were used as chelators for the preparation of new Cu(i) phosphane complexes 1-4. They were synthesized by the reaction of [Cu(CH3CN)4]PF6 and triphenylphosphine or 1,3,5-triaza-7-phosphaadamantane with LOiPr and L2OiPr ligands, in acetonitrile or acetonitrile/methanol solution. The authenticity of the compounds was confirmed by CHN analysis, 1H-, 13C- and 31P-NMR, FT-IR spectroscopy, and electrospray ionization mass spectrometry (ESI-MS). Furthermore, the electronic and molecular structures of the selected Cu(i) coordination compound 3 were investigated by synchrotron radiation-induced X-ray photoelectron spectroscopy (SR-XPS), and the local structure around the copper ion site was studied combining X-ray absorption fine structure (XAFS) spectroscopy techniques and DFT modelling. Triphenylphosphine as a coligand confers to [Cu(LOiPr)(PPh3)]PF6 (1) and [Cu(L2OiPr)(PPh3)]PF6 (3) a significant antitumor activity in 3D spheroidal models of human colon cancer cells. Investigations focused on the mechanism of action evidenced protein disulfide-isomerase (PDI) as an innovative molecular target for this class of phosphane copper(i) complexes. By hampering PDI activity, copper(i) complexes were able to cause an imbalance in cancer cell redox homeostasis thus leading to cancer cell death - a non-apoptotic programmed cell death.
Collapse
Affiliation(s)
- Maura Pellei
- School of Science and Technology, Chemistry Division, University of Camerino Via Madonna delle Carceri (ChIP) 62032 Camerino Italy
| | - Carlo Santini
- School of Science and Technology, Chemistry Division, University of Camerino Via Madonna delle Carceri (ChIP) 62032 Camerino Italy
| | - Miriam Caviglia
- School of Science and Technology, Chemistry Division, University of Camerino Via Madonna delle Carceri (ChIP) 62032 Camerino Italy
| | - Jo' Del Gobbo
- School of Science and Technology, Chemistry Division, University of Camerino Via Madonna delle Carceri (ChIP) 62032 Camerino Italy
| | - Chiara Battocchio
- Department of Science, Roma Tre University Via della Vasca Navale 79 00146 Roma Italy
| | - Carlo Meneghini
- Department of Science, Roma Tre University Via della Vasca Navale 79 00146 Roma Italy
| | - Simone Amatori
- Department of Science, Roma Tre University Via della Vasca Navale 79 00146 Roma Italy
| | - Chiara Donati
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova Via Marzolo 5 35131 Padova Italy
| | - Eleonora Zampieri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova Via Marzolo 5 35131 Padova Italy
| | - Valentina Gandin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova Via Marzolo 5 35131 Padova Italy
| | - Cristina Marzano
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova Via Marzolo 5 35131 Padova Italy
| |
Collapse
|
22
|
Zhang H. A focus on copper depletion-induced cuproptosis for cancer therapy. Chem Sci 2025:d5sc90034d. [PMID: 39975765 PMCID: PMC11834195 DOI: 10.1039/d5sc90034d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025] Open
Abstract
Copper has emerged as a promising target for cancer therapy, with extensive studies on copper accumulation-induced cuproptosis. However, the potential of copper depletion-induced cuproptosis remains largely unexplored. Recently, Zhou et al. (M. Zhou, F. Muhammad, Y. Zhang, T. Li, J. Feng, J. Zhao and H. Wei, Chem. Sci., 2025, https://doi.org/10.1039/D4SC04712E) reported an innovative strategy for copper depletion-based cuproptosis. Notably, this approach leverages the solubility product principle, a mechanism not previously addressed in studies, to achieve effective tumor therapy through the disruption of copper homeostasis.
Collapse
Affiliation(s)
- Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Jilin Changchun 130022 China
- Department of Chemistry, Tsinghua University Beijing 100084 China
| |
Collapse
|
23
|
Wang C, Chen D, Wei Z, Tan J, Wu C, Zhang X. Metal-Catalyzed Abiotic Cleavage of C═C Bonds for Effective Fluorescence Imaging of Cu(II) and Fe(III) in Living Systems. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412407. [PMID: 39784410 PMCID: PMC11848571 DOI: 10.1002/advs.202412407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 12/25/2024] [Indexed: 01/12/2025]
Abstract
Imaging abnormal copper/iron with effective fluorescent tools is essential to comprehensively put insight into many pathological events. However, conventional coordination-based detection is mired in the fluorescence quenching induced by paramagnetic Cu(II)/Fe(III). Moreover, the strong chelating property of the probe will consume dissociative metal ions and inevitably interfere with the physiological microenvironment. Here, a new strategy is developed by employing this aberrant Cu(II)/Fe(III) to catalyze bond cleavage for fluorescent imaging of them. A short series of near-infrared fluorescent molecules (NIRB1-NIRB6) is devised as substrates, wherein the specific C═C bonds can be effectively cleaved to activate red fluorophore by Cu(II)/Fe(III) catalyzing. Representatively, NIRB1 is applied for fluorescent imaging of Cu(II)/Fe(III) in living cells, zebrafish, and Alzheimer's disease (AD)-afflicted mouse brains which is of significance to monitor metal safety. The successful cleavage of C═C bonds catalyzed by Cu(II)/Fe(III) enriches the application of abiotic bond cleavage reactions in metal detection, and may also inspire the development of fluorescent tools for the future diagnosis and therapy of diseases.
Collapse
Affiliation(s)
- Chunfei Wang
- Faculty of Health SciencesUniversity of MacauMacau SAR999078China
- Department of PharmacologySchool of PharmacyWannan Medical CollegeWuhuAnhui241002China
| | - Dandan Chen
- Department of Biomedical EngineeringSouthern University of Science and TechnologyShenzhenGuangdong518055China
| | - Zixiang Wei
- Faculty of Health SciencesUniversity of MacauMacau SAR999078China
| | - Jingyun Tan
- Faculty of Health SciencesUniversity of MacauMacau SAR999078China
| | - Changfeng Wu
- Department of Biomedical EngineeringSouthern University of Science and TechnologyShenzhenGuangdong518055China
| | - Xuanjun Zhang
- Faculty of Health SciencesUniversity of MacauMacau SAR999078China
- MOE Frontiers Science Centre for Precision OncologyUniversity of MacauMacau SAR999078China
| |
Collapse
|
24
|
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 2025; 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] [MESH Headings] [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
|
25
|
Ganapathy V, Jaganathan R, Chinnaiyan M, Chengizkhan G, Sadhasivam B, Manyanga J, Ramachandran I, Queimado L. E-Cigarette effects on oral health: A molecular perspective. Food Chem Toxicol 2025; 196:115216. [PMID: 39736445 PMCID: PMC11976636 DOI: 10.1016/j.fct.2024.115216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/09/2024] [Accepted: 12/21/2024] [Indexed: 01/01/2025]
Abstract
Electronic cigarettes (e-cigarettes) have emerged as a potential alternative to traditional smoking and may aid in tobacco harm reduction and smoking cessation. E-cigarette use has notably increased, especially among young non-tobacco users, raising concerns due to the unknown long-term health effects. The oral cavity is the first and one of the most crucial anatomical sites for the deposition of e-cigarette aerosols. E-cigarette aerosols contain nicotine, flavors, volatile organic compounds, heavy metals, carcinogens, and other hazardous substances. These aerosols impact the oral cavity, disrupting host-microbial interactions and triggering gingivitis and systemic diseases. Furthermore, oral inflammation and periodontitis can be caused by proinflammatory cytokines induced by e-cigarette aerosols. The toxic components of e-cigarette aerosols increase the cellular reactive oxygen species (ROS) levels, reduce antioxidant capacity, increase DNA damage, and disrupt repair processes, which may further contribute to harmful effects on oral epithelum, leading to inflammatory and pre-malignant oral epithelial lesions. In this review, we analyze the toxicological properties of compounds in e-cigarette aerosols, exploring their cytotoxic, genotoxic, and inflammatory effects on oral health and delving into the underlying molecular mechanisms. Further research is essential to understand the impact of e-cigarettes on oral health and make informed regulatory decisions based on reliable scientific evidence.
Collapse
Affiliation(s)
- Vengatesh Ganapathy
- Department of Otolaryngology-Head and Neck Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Ravindran Jaganathan
- Preclinical Department, Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL-RCMP), Ipoh, Perak, 30450, Malaysia
| | - Mayilvanan Chinnaiyan
- Department of Otolaryngology-Head and Neck Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Gautham Chengizkhan
- Department of Otolaryngology-Head and Neck Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Balaji Sadhasivam
- Department of Otolaryngology-Head and Neck Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Occupational and Environmental Health, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Jimmy Manyanga
- Department of Otolaryngology-Head and Neck Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Ilangovan Ramachandran
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600113, Tamil Nadu, India
| | - Lurdes Queimado
- Department of Otolaryngology-Head and Neck Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; TSET Health Promotion Research Center, Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
26
|
Oliva CR, Ali MY, Flor S, Griguer CE. Copper-Induced Enhancement of Glioblastoma Tumorigenicity via Cytochrome C Oxidase. Antioxidants (Basel) 2025; 14:142. [PMID: 40002329 PMCID: PMC11851629 DOI: 10.3390/antiox14020142] [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: 12/20/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Copper is an essential trace element, yet chronic copper exposure can lead to toxicity in humans, and high levels of copper have been found in the blood or tumors of patients with various forms of cancer and may affect cancer severity and response to treatment. Copper is required for the activation of cytochrome c oxidase (CcO), the mitochondrial complex that facilitates oxidative phosphorylation (OXPHOS)-mediated ATP production. We recently reported that the increased activation of CcO underlies the acquisition of treatment resistance in glioblastoma (GBM) cells. However, the potential role of copper in GBM progression or treatment resistance has not been investigated. Here, we present evidence that exposure to 20 µM copper, the maximum allowable limit for public water supplies set by the U.S. Environmental Protection Agency, promotes GBM tumor growth and reduces overall survival in vivo and increases GBM cell resistance to radiation and chemotherapy in vitro. In vitro exposure to 20 µM copper substantially increased the activity of CcO, elevated the rate and level of ATP production, and triggered a metabolic shift to an OXPHOS phenotype in GBM cells. Furthermore, copper exposure led to a substantial increase in the accumulation of glutathione and glutathione precursors in these cells. These findings establish copper as a tumor promoter in GBM and suggest that copper mediates these effects through the upregulation of CcO activity, which enhances OXPHOS metabolism and glutathione production.
Collapse
Affiliation(s)
- Claudia R. Oliva
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa, IA 52242, USA; (C.R.O.); (M.Y.A.); (S.F.)
| | - Md Yousuf Ali
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa, IA 52242, USA; (C.R.O.); (M.Y.A.); (S.F.)
- Mass General Hospital Center for Cancer Research, Harvard Medical School, Boston, MA 02129, USA
| | - Susanne Flor
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa, IA 52242, USA; (C.R.O.); (M.Y.A.); (S.F.)
| | - Corinne E. Griguer
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa, IA 52242, USA; (C.R.O.); (M.Y.A.); (S.F.)
| |
Collapse
|
27
|
Pastuch-Gawołek G, Szreder J. Effect of Glycoconjugation on Cytotoxicity and Selectivity of 8-Aminoquinoline Derivatives Compared to 8-Hydroxyquinoline. Molecules 2025; 30:427. [PMID: 39860296 PMCID: PMC11767929 DOI: 10.3390/molecules30020427] [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: 12/21/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Numerous emerging chemotherapeutic agents incorporate N-heterocyclic fragments in their structures, with the quinoline skeleton being particularly significant. Our recent works have focused on glycoconjugates of 8-hydroxyquinoline (8-HQ), which demonstrated enhanced bioavailability and solubility compared to their parent compounds, although they fell short in selectivity. In this study, our objective was to improve the selectivity of glycoconjugates by replacing the oxygen atom with nitrogen by substituting the 8-HQ moiety with 8-aminoquinoline (8-AQ). The 8-AQ derivatives were functionalized through the amino group and linked to sugar derivatives (D-glucose or D-galactose) that were modified with an azide, alkylazide, or propargyl group at the anomeric position by copper(I)-catalyzed 1,3-dipolar azido-alkyne cycloaddition (CuAAC). The resulting glycoconjugates, as well as their potential metabolites, were evaluated for their ability to inhibit the proliferation of cancer cell lines (including HCT 116 and MCF-7) and a healthy cell line (NHDF-Neo). Two of the synthesized glycoconjugates (17 and 18) demonstrated higher cytotoxicity than their oxygen-containing counterparts and showed improved selectivity for cancer cells, thus enhancing their anticancer potential. Furthermore, it was found that glycoconjugates exhibited greater cytotoxicity in comparison to their potential metabolites.
Collapse
Affiliation(s)
- Gabriela Pastuch-Gawołek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland;
- Biotechnology Center, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| | - Julia Szreder
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland;
| |
Collapse
|
28
|
Wang W, Lu D, Yang H, Chen Z, Ling W, Song S, Peng L, Liu Q, Jiang G. Unveiling the Origin of Copper Accumulation in Plasma with Aging. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:58-67. [PMID: 39839250 PMCID: PMC11744393 DOI: 10.1021/envhealth.4c00096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 01/23/2025]
Abstract
Aging is intricately linked to various diseases including cancers, neurodegenerative disorders, and metabolic irregularities. Copper (Cu) overexposure has been found to be linked to many diseases during aging, particularly neurodegenerative diseases. Meanwhile, as an essential element, Cu has been implicated in key processes associated with aging, raising questions about its role in age-related health issues. This study delves into the mechanisms behind the copper imbalance during aging. By analyzing blood copper concentrations of healthy individuals (including data from healthy subjects (26 ≤ age ≤ 90, n = 62) and publicly available data from the National Health and Nutrition Examination Survey (18 ≤ age < 80, n = 1624)) and employing C57BL/6N male mice models (n = 22), we reveal a consistent age-related increase in copper levels, particularly in plasma. Utilizing stable copper isotopic analysis, copper-associated protein analysis, and metabolomic analysis, we trace the sources of Cu imbalance associated with aging. Our findings reveal that aged mice had a higher copper concentrations and an enrichment of light copper isotope (63Cu) in plasma compared to controls. Additionally, copper reductases and copper transporters are upregulated in the intestine tract, associated with the AMPK and mTOR signaling pathways. We suggest that aged mice have an abnormally high copper intake requirement, probably due to deregulated nutrient sensing, leading to increased expression levels of copper reductases and copper transporters for extra copper absorption in the intestines. This research provides a copper-centric perspective on the connection between deregulated nutrient sensing and aging, thus shedding light on the aspect of aging and copper overexposure.
Collapse
Affiliation(s)
- Weichao Wang
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- School
of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
| | - Dawei Lu
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Hang Yang
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Zigu Chen
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Weibo Ling
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Shanjun Song
- National
Institute of Metrology, Beijing 100029, China
| | - Linyi Peng
- Department
of Rheumatology & Clinical Immunology, National Clinical Research
Center for Dermatologic & Immunologic Diseases (NCRC-DID), Peking
Union Medical College Hospital, Chinese
Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Qian Liu
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- Institute
of Environment and Health, Jianghan University, Wuhan 430056, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Guibin Jiang
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- School
of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
| |
Collapse
|
29
|
Falcone E, Vigna V, Schueffl H, Stellato F, Vileno B, Bouraguba M, Mazzone G, Proux O, Morante S, Heffeter P, Sicilia E, Faller P. When Metal Complexes Evolve, and a Minor Species is the Most Active: the Case of Bis(Phenanthroline)Copper in the Catalysis of Glutathione Oxidation and Hydroxyl Radical Generation. Angew Chem Int Ed Engl 2025; 64:e202414652. [PMID: 39363702 PMCID: PMC11720388 DOI: 10.1002/anie.202414652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
Several copper-ligands, including 1,10-phenanthroline (Phen), have been investigated for anticancer purposes based on their capacity to bind excess copper (Cu) in cancer tissues and form redox active complexes able to catalyse the formation of reactive oxygen species (ROS), ultimately leading to oxidative stress and cell death. Glutathione (GSH) is a critical compound as it is highly concentrated intracellularly and can reduce and dissociate copper(II) from the ligand forming poorly redox-active copper(I)-thiolate clusters. Here we report that Cu-Phen2 speciation evolves in physiologically relevant GSH concentrations. Experimental and computational experiments suggest that at pH 7.4 mostly copper(I)-GSH clusters are formed, but a minor species of copper(I) bound to one Phen and forming ternary complexes with GSH (GS-Cu-Phen) is the redox active species, oxidizing quite efficiently GSH to GSSG and forming HO⋅ radicals. This minor active species becomes more populated at lower pH, such as typical lysosomal pH 5, resulting in faster GSH oxidation and HO⋅ production. Consistently, cell culture studies showed lower toxicity of Cu-Phen2 upon inhibition of lysosomal acidification. Overall, this study underscores that sub-cellular localisation can considerably influence the speciation of Cu-based drugs and that minor species can be the most redox- and biologically-active.
Collapse
Affiliation(s)
- Enrico Falcone
- Institut de Chimie (UMR 7177)University of Strasbourg, CNRS4 Rue Blaise Pascal67081StrasbourgFrance
- current address: Laboratoire de Chimie de Coordination (UPR 8142)CNRS31077ToulouseFrance
| | - Vincenzo Vigna
- Department of Chemistry and Chemical TechnologiesUniversità della Calabria87036Arcavacata di RendeCSItaly
| | - Hemma Schueffl
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of Vienna1090ViennaAustria
| | - Francesco Stellato
- Department of PhysicsUniversità di Roma Tor VergataVia della Ricerca Scientifica 100133RomaItaly
- INFNUniversità di Roma Tor VergataVia della Ricerca Scientifica 100133RomaItaly
| | - Bertrand Vileno
- Institut de Chimie (UMR 7177)University of Strasbourg, CNRS4 Rue Blaise Pascal67081StrasbourgFrance
| | - Merwan Bouraguba
- Institut de Chimie (UMR 7177)University of Strasbourg, CNRS4 Rue Blaise Pascal67081StrasbourgFrance
| | - Gloria Mazzone
- Department of Chemistry and Chemical TechnologiesUniversità della Calabria87036Arcavacata di RendeCSItaly
| | - Olivier Proux
- Observatoire des Sciences de l'Univers de Grenoble, UAR 832CNRS-Université Grenoble Alpes38041GrenobleFrance
| | - Silvia Morante
- Department of PhysicsUniversità di Roma Tor VergataVia della Ricerca Scientifica 100133RomaItaly
- INFNUniversità di Roma Tor VergataVia della Ricerca Scientifica 100133RomaItaly
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of Vienna1090ViennaAustria
| | - Emilia Sicilia
- Department of Chemistry and Chemical TechnologiesUniversità della Calabria87036Arcavacata di RendeCSItaly
| | - Peter Faller
- Institut de Chimie (UMR 7177)University of Strasbourg, CNRS4 Rue Blaise Pascal67081StrasbourgFrance
- Institut Universitaire de France (IUF)1 rue Descartes75231ParisFrance
| |
Collapse
|
30
|
Xia Z, Zong S, Zhou S, Rao Y, Li Z. Rheumatoid arthritis and gliostatin: a two-sample Mendelian randomization analysis and RT-PCR validation. Clin Rheumatol 2025; 44:125-132. [PMID: 39352437 DOI: 10.1007/s10067-024-07164-3] [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: 07/30/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 01/14/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease of unknown etiology. Cuproptosis, a novel form of cell death, is characterized by cytotoxicity originating from copper ions. To date, the relationship between cuproptosis-related gene gliostatin (GLS) and RA. METHODS All raw data were retrieved from the Gene Expression Omnibus (GEO) public database. The expression level of genes between RA and healthy samples was evaluated to identify differentially expressed genes. Then, LASSO regression was used to screen disease signature genes, and a nomogram was constructed based on five hub genes to predict disease scores. Validation experiments were performed using quantitative real-time PCR (qRT-PCR) to detect the most significant CRGs. Finally, the causal relationship between GLS and RA was analyzed through Mendelian randomization methodology. RESULTS Five differentially expressed CRGs (NLRP3, ATP7A, MTF1, GLS, and DBT) were identified between normal and RA samples, all of which were validated as disease-specific genes through LASSO regression analysis. Meanwhile, the nomogram demonstrated a positive correlation between RA and the expression of GLS. Furthermore, q-PCR revealed that the expression level of GLS was higher in RA patients compared to those in the control group. Taken together, a causal relationship between GLS and RA was corroborated through Mendelian randomization. CONCLUSION GLS, a cuproptosis-related gene, is closely associated with RA and plays a significant role in its diagnosis. Key Points • The causal relationship between GLS and RA is proved by Mendelian randomization.
Collapse
Affiliation(s)
- Zhongbin Xia
- Health Management Medicine Department, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Siwen Zong
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Sibin Zhou
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yujie Rao
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhenjiang Li
- Department of Hematology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
31
|
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
|
32
|
Zeng T, Lei GL, Yu ML, Zhang TY, Wang ZB, Wang SZ. The role and mechanism of various trace elements in atherosclerosis. Int Immunopharmacol 2024; 142:113188. [PMID: 39326296 DOI: 10.1016/j.intimp.2024.113188] [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: 06/28/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
Atherosclerosis is a slow and complex disease that involves various factors, including lipid metabolism disorders, oxygen-free radical production, inflammatory cell infiltration, platelet adhesion and aggregation, and local thrombosis. Trace elements play a crucial role in human health. Many trace elements, especially metallic ones, not only maintain the normal functions of organs but also participate in basic metabolic processes. The latest studies have revealed a close correlation between trace elements and the occurrence and progression of atherosclerosis. The imbalance of these trace elements can induce atherosclerosis or accelerate its progression through various mechanisms, which poses a significant threat to human health. Therefore, exploring the specific mechanism of trace elements on atherosclerosis is highly significant. In this review, we summarized the roles and mechanisms of iron, copper, zinc, magnesium, and selenium homeostasis and imbalance in atherosclerosis development, in order to identify novel targets and therapeutic strategies for treating atherosclerosis.
Collapse
Affiliation(s)
- Tao Zeng
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Guan-Lan Lei
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Mei-Ling Yu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Ting-Yu Zhang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Zong-Bao Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| |
Collapse
|
33
|
Hu J, Zhu J, Chen T, Zhao Y, Xu Q, Wang Y. Cuproptosis in cancer therapy: mechanisms, therapeutic application and future prospects. J Mater Chem B 2024; 12:12191-12206. [PMID: 39526989 DOI: 10.1039/d4tb01877j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Cuproptosis is a regulated form of cell death induced by the accumulation of metal ions and is closely linked to aspects of cellular drug resistance, cellular metabolism, and signalling pathways. Due to its crucial role in regulating physiological and pathological processes, cuproptosis has gained increasing significance as a potential target for anticancer drug development. In this review, we introduce the definition of cuproptosis and provide a comprehensive discussion of the mechanisms of cuproptosis. In addition, the methods for the detection of cuproptosis are summarized, and recent advances in cuproptosis in cancer therapy are reviewed, mainly in terms of elesclomol (ES)-mediated cuproptosis and disulfiram (DSF)-mediated cuproptosis, which provided practical value for applications. Finally, the current challenges and future development of cuproptosis-mediated cancer therapy are discussed. In summary, this review highlights recent progress on cuproptosis in cancer therapy, offering novel ideas and strategies for future research and applications.
Collapse
Affiliation(s)
- Jiawei Hu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Junfei Zhu
- China-Japan Friendship Hospital, No. 2 Sakura East Street, Chaoyang District, Beijing, China
| | - Tao Chen
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yudie Zhao
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Qingwen Xu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| |
Collapse
|
34
|
Wang K, Wang L, Shang Z, Yang X, Li H, Wang X, Zhu M, Meng Q. A series of DNA targeted Cu (II) complexes containing 1,8-naphthalimide ligands: Synthesis, characterization and in vitro anticancer activity. J Inorg Biochem 2024; 261:112721. [PMID: 39236444 DOI: 10.1016/j.jinorgbio.2024.112721] [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: 06/21/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024]
Abstract
Copper(II) complexes are very promising candidates for platinum-based anticancer agents. Herein, three Cu (II) complexes (1-3) containing 1,8-naphthalimide ligands were synthesized and characterized by FT-IR, elemental analysis, ESI-MS and single crystal X-ray diffraction (complex 3). In addition, a control compound (complex 4) without 1,8-naphthalimide ligand was synthesized and characterized. The in vitro anticancer activity of the synthesized complexes against five cancer cell lines and one normal cell line was evaluated by MTS assay. The results displayed the antitumor activity of complexes 1-3 was controlled by the aliphatic chain length of ligands, their cytotoxicity was in the order 3 > 2 > 1, giving the IC50 values ranging from 2.874 ± 0.155 μM to 31.47 ± 0.29 μM against five cancer cell lines. Complex 4 showed less activity in comparison with complex 1-3. Notably, complexes 1-3 displayed much higher selectivity (SI = 2.65 to 10.16) compared to complex 4 (SI = 1.0), indicated that the introduction of 1,8-naphthalimide group not only increased the activity of this series of compounds but also enhanced their specific selectivity to cancer cells. Compound 3 induced apoptosis in cancer cells and blocked the S-phase and G2/M of cancer cells. The interaction with DNA of complexes 3 and 4 was studied by UV/Vis spectroscopic titrations, competitive DNA-binding experiment, viscometry and CD spectra. The results showed that complex 3 interacted with DNA in an intercalating mode, but the interaction mode of compound 4 with DNA was electrostatic interaction.
Collapse
Affiliation(s)
- Kehua Wang
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan, Liaoning 114051, PR China; Liaoning Key Laboratory of Development and Utilization for Natural Products Active Molecules, Anshan Normal University, Anshan, Liaoning 114007, PR China
| | - Ling Wang
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Zhuye Shang
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan, Liaoning 114051, PR China
| | - Xingzhi Yang
- Center for Natural Drug Activity Screening, Kunming Institute of Botany, Chinese Academy of Sciences, PR China
| | - Hongmei Li
- Center for Natural Drug Activity Screening, Kunming Institute of Botany, Chinese Academy of Sciences, PR China
| | - Xiaochun Wang
- Liaoning Key Laboratory of Development and Utilization for Natural Products Active Molecules, Anshan Normal University, Anshan, Liaoning 114007, PR China
| | - Mingchang Zhu
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Qingtao Meng
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan, Liaoning 114051, PR China.
| |
Collapse
|
35
|
El Yaagoubi OM, Oularbi L, Salhi O, Samaki H, El Rhazi M, Aboudkhil S. Novel copper complex inhibits the proteasome in skin squamous cell carcinoma induced by DMBA in mice. J Trace Elem Med Biol 2024; 86:127533. [PMID: 39321648 DOI: 10.1016/j.jtemb.2024.127533] [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: 04/14/2024] [Revised: 08/11/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
The proteasomal system is becoming a target for the treatment of several diseases, especially in cancer therapy. The present study aims to develop a novel copper complex that inhibits the proteasome in skin squamous cell carcinoma. New molecules based on the copper complex were synthesized for the first time to assess their potential as proteasome inhibitors, specifically targeting squamous cell carcinoma induced by 7,12-dimethylbenz(a)anthracene (DMBA) in mouse models. Fourier Transform Infrared (FTIR), X-ray diffraction (XRD), scanning electron microscopy (SEM), nuclear magnetic resonance (NMR), and energy dispersive X-ray analysis (EDX) were carried out to characterize this new copper complex. Notably, the presence of a papilloma (skin tumor) was confirmed by histopathological analysis. Subsequent investigation included the quantification of proteasome levels using a sandwich ELISA test, and the catalytic activity of the 20S proteasome was determined by measuring the fluorescence emitted after the cleavage of 7-amino-4-methylcoumarin (AMC). Hence, X-ray crystallography indicates that all Cu atoms are five-coordinated in a square-pyramidal configuration and biological activity of copper Schiff base complex, which exhibits high proteasome inhibitory activities with particular selectivity of β5 subunit. The pharmacokinetic properties (ADMET) of the copper complex named Cu(L1) showed encouraging results with very low toxicity, distribution, and absorption. Structure-activity relationship (SAR) information obtained from Cu(L1) demonstrated its selectivity and potent inhibition for β5 subunit. In this regard, this copper complex has emerged as a novel therapy for skin cancer.
Collapse
Affiliation(s)
- Ouadie Mohamed El Yaagoubi
- Laboratory of Biochemistry, Environment and Agri-Food (URAC 36), Faculty of Sciences and Techniques - Mohammedia, Hassan II University, Casablanca, Morocco.
| | - Larbi Oularbi
- Laboratory of Materials Membranes and Environment, P.B 146, Faculty of Sciences and Techniques - Mohammedia, Hassan II University, Casablanca, Morocco; Supramolecular Nanomaterials Group (SNG), Mohammed VI Polytechnic University, Benguerir, Morocco.
| | - Ouissal Salhi
- Laboratory of Materials Membranes and Environment, P.B 146, Faculty of Sciences and Techniques - Mohammedia, Hassan II University, Casablanca, Morocco.
| | - Hamid Samaki
- National Institute of Social Action (INAS), Tangier, Morocco.
| | - Mama El Rhazi
- Laboratory of Materials Membranes and Environment, P.B 146, Faculty of Sciences and Techniques - Mohammedia, Hassan II University, Casablanca, Morocco.
| | - Souad Aboudkhil
- Laboratory of Biochemistry, Environment and Agri-Food (URAC 36), Faculty of Sciences and Techniques - Mohammedia, Hassan II University, Casablanca, Morocco.
| |
Collapse
|
36
|
Bhatnagar K, Jha K, Dalal N, Patki N, Gupta G, Kumar A, Kumar A, Chaudhary S. Exploring micronutrients and microbiome synergy: pioneering new paths in cancer therapy. Front Immunol 2024; 15:1442788. [PMID: 39676876 PMCID: PMC11638209 DOI: 10.3389/fimmu.2024.1442788] [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: 06/02/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
The human microbiome is the complex ecosystem consisting of trillions of microorganisms that play a key role in developing the immune system and nutrient metabolism. Alterations in the gut microbiome have been linked to cancer initiation, progression, metastasis, and response to treatment. Accumulating evidence suggests that levels of vitamins and minerals influence the gut environment and may have implications for cancer risk and progression. Bifidobacterium has been reported to reduce the colorectal cancer risk by binding to free iron. Additionally, zinc ions have been shown to activate the immune cells and enhance the effectiveness of immunotherapy. Higher selenium levels have been associated with a reduced risk of several cancers, including colorectal cancer. In contrast, enhanced copper uptake has been implicated in promoting cancer progression, including colon cancer. The interaction between cancer and gut bacteria, as well as dysbiosis impact has been studied in animal models. The interplay between prebiotics, probiotics, synbiotics, postbiotics and gut bacteria in cancer offers the diverse physiological benefits. We also explored the particular probiotic formulations like VSL#3, Prohep, Lactobacillus rhamnosus GG (LGG), etc., for their ability to modulate immune responses and reduce tumor burden in preclinical models. Targeting the gut microbiome through antibiotics, bacteriophage, microbiome transplantation-based therapies will offer a new perspective in cancer research. Hence, to understand this interplay, we outline the importance of micronutrients with an emphasis on the immunomodulatory function of the microbiome and highlight the microbiome's potential as a target for precision medicine in cancer treatment.
Collapse
Affiliation(s)
- Kartik Bhatnagar
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Kanupriya Jha
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Nishu Dalal
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Ninad Patki
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Garima Gupta
- Biological Engineering and Sciences, Indian Institute of Technology Gandhinagar Palaj, Gandhinagar, Gujarat, India
| | - Amit Kumar
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
| | - Sarika Chaudhary
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
37
|
Gao H, Chen Z, Yao Y, He Y, Hu X. Common biological processes and mutual crosstalk mechanisms between cardiovascular disease and cancer. Front Oncol 2024; 14:1453090. [PMID: 39634266 PMCID: PMC11614734 DOI: 10.3389/fonc.2024.1453090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Cancer and cardiovascular disease (CVD) are leading causes of mortality and thus represent major health challenges worldwide. Clinical data suggest that cancer patients have an increased likelihood of developing cardiovascular disease, while epidemiologic studies have shown that patients with cardiovascular disease are also more likely to develop cancer. These observations underscore the increasing importance of studies exploring the mechanisms underlying the interaction between the two diseases. We review their common physiological processes and potential pathophysiological links. We explore the effects of chronic inflammation, oxidative stress, and disorders of fatty acid metabolism in CVD and cancer, and also provide insights into how cancer and its treatments affect heart health, as well as present recent advances in reverse cardio-oncology using a new classification approach.
Collapse
Affiliation(s)
- Hanwei Gao
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Zhongyu Chen
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| | - Yutong Yao
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| | - Yuquan He
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| | - Xin Hu
- Department of Cardiology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- CJUH-JLU-China iGEM Team, Jilin University, Changchun, Jilin, China
| |
Collapse
|
38
|
Guo N, Li X, Xie L, Hao S, Zhou X. A quantitative review of the effects of biochar application on the reduction of Cu concentration in plant: a meta-analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:60731-60748. [PMID: 39392574 DOI: 10.1007/s11356-024-34789-3] [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: 03/07/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024]
Abstract
Contamination and toxicity of copper (Cu) in soils are global issues, particularly in regions where Cu-based fungicides are utilized. Elevated Cu concentrations can lead to soil contamination and pose significant risks to the ecosystem, including plant life, wildlife, and human health. The application of biochar has been proposed as a viable strategy to mitigate Cu accumulation in plants. However, there is no quantitative and data-based consensus on the impact of biochar on plant Cu accumulation. In this meta-analysis, 624 data records from 65 published literature were collected and the effects of various factors, including biochar properties, experimental conditions, and soil properties on Cu accumulation in plants, were examined through meta-subgroup analysis and meta-regression models. The results obtained indicate a significant dose-dependent effect of biochar in decreasing Cu concentration in plants by an average of 23.45%. Soils with acidic pH values and medium textures were more conducive for biochar to mitigate Cu accumulation in plant tissues. In addition, manure biochar and green waste biochar were found to be more successful in decreasing Cu concentrations in plants compared to other biochar types. Biochar types with pyrolysis temperatures of > 600 °C and pH values of ≥ 10 resulted in greater decreases in plant Cu concentration. Regarding biochar application, biochar minimum range of 1% in potting experiments and 20 t/ha in field experiments have been recommended to effectively decrease Cu concentration in plants. Overall, these findings provide valuable insights into Cu transfer mitigation through food chain to human bodies and for policymakers to take preventive measures.
Collapse
Affiliation(s)
- Ningyu Guo
- College of Resources and Environment, Southwest University, Chongqing, 400715, China
| | - Xue Li
- College of Resources and Environment, Southwest University, Chongqing, 400715, China
| | - Linzhi Xie
- College of Resources and Environment, Southwest University, Chongqing, 400715, China
| | - Shangyan Hao
- College of Resources and Environment, Southwest University, Chongqing, 400715, China
| | - Xinbin Zhou
- College of Resources and Environment, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
39
|
Fitisemanu FM, Padilla-Benavides T. Emerging perspectives of copper-mediated transcriptional regulation in mammalian cell development. Metallomics 2024; 16:mfae046. [PMID: 39375833 PMCID: PMC11503025 DOI: 10.1093/mtomcs/mfae046] [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: 06/23/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Copper (Cu) is a vital micronutrient necessary for proper development and function of mammalian cells and tissues. Cu mediates the function of redox active enzymes that facilitate metabolic processes and signaling pathways. Cu levels are tightly regulated by a network of Cu-binding transporters, chaperones, and small molecule ligands. Extensive research has focused on the mammalian Cu homeostasis (cuprostasis) network and pathologies, which result from mutations and perturbations. There are roles for Cu-binding proteins as transcription factors (Cu-TFs) and regulators that mediate metal homeostasis through the activation or repression of genes associated with Cu handling. Emerging evidence suggests that Cu and some Cu-TFs may be involved in the regulation of targets related to development-expanding the biological roles of Cu-binding proteins. Cu and Cu-TFs are implicated in embryonic and tissue-specific development alongside the mediation of the cellular response to oxidative stress and hypoxia. Cu-TFs are also involved in the regulation of targets implicated in neurological disorders, providing new biomarkers and therapeutic targets for diseases such as Parkinson's disease, prion disease, and Friedreich's ataxia. This review provides a critical analysis of the current understanding of the role of Cu and cuproproteins in transcriptional regulation.
Collapse
|
40
|
Pandey P, Ramniwas S, Pandey S, Lakhanpal S, Padmapriya G, Mishra S, Kaur M, Ashraf A, Kumar MR, Khan F. Review to Elucidate the Correlation between Cuproptosis-Related Genes and Immune Infiltration for Enhancing the Detection and Treatment of Cervical Cancer. Int J Mol Sci 2024; 25:10604. [PMID: 39408933 PMCID: PMC11477161 DOI: 10.3390/ijms251910604] [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: 09/09/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Copper is a vital trace element in oxidized and reduced forms. It plays crucial roles in numerous biological events such as redox chemistry, enzymatic reactions, mitochondrial respiration, iron metabolism, autophagy, and immune modulation. Maintaining the balance of copper in the body is essential because its deficiency and excess can be harmful. Abnormal copper metabolism has a two-fold impact on the development of tumors and cancer treatment. Cuproptosis is a form of cell death that occurs when there is excessive copper in the body, leading to proteotoxic stress and the activation of a specific pathway in the mitochondria. Research has been conducted on the advantageous role of copper ionophores and chelators in cancer management. This review presents recent progress in understanding copper metabolism, cuproptosis, and the molecular mechanisms involved in using copper for targeted therapy in cervical cancer. Integrating trace metals and minerals into nanoparticulate systems is a promising approach for controlling invasive tumors. Therefore, we have also included a concise overview of copper nanoformulations targeting cervical cancer cells. This review offers comprehensive insights into the correlation between cuproptosis-related genes and immune infiltration, as well as the prognosis of cervical cancer. These findings can be valuable for developing advanced clinical tools to enhance the detection and treatment of cervical cancer.
Collapse
Affiliation(s)
- Pratibha Pandey
- Post Doctoral Department, Eudoxia Research University, New Castle, DE 19808, USA;
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, India
| | - Seema Ramniwas
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India;
| | - Shivam Pandey
- School of Applied and Life Sciences, Uttaranchal University, Dehradun 248007, India;
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India;
| | - G. Padmapriya
- Department of Chemistry and Biochemistry, School of Sciences, JAIN Deemed to be University, Bangalore 560069, India;
| | - Shivang Mishra
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur 303121, India;
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur 303012, India;
| | - Ayash Ashraf
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali 140307, India;
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam 531162, India;
| | - Fahad Khan
- Center for Global Health Research Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, India
| |
Collapse
|
41
|
Chen Y, Liao Y, Huang L, Luo Z. Exploring copper metabolism-induced cell death in gastric cancer: a single-cell RNA sequencing study and prognostic model development. Discov Oncol 2024; 15:482. [PMID: 39331287 PMCID: PMC11436710 DOI: 10.1007/s12672-024-01374-6] [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: 08/14/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the third leading cause of cancer-related deaths globally. Despite advancements in treatment, the overall 5-year survival rate remains below 30%, particularly in advanced stages. Copper metabolism, vital for various cellular processes, has been linked to cancer progression, but its role in GC, especially at the single-cell level, is not well understood. OBJECTIVE This study aims to investigate copper metabolism in GC by integrating single-cell RNA sequencing (scRNA-seq) data and developing a prognostic model based on copper metabolism-related gene (CMRG) expression. The study explores how copper metabolism affects the tumor microenvironment and identifies potential therapeutic targets. METHODS scRNA-seq data from gastric cancer and normal tissues were analyzed using the Seurat package. Principal Component Analysis (PCA) and Uniform Manifold Approximation and Projection (UMAP) were used for dimensionality reduction and clustering. Non-negative matrix factorization (NMF) was employed for T cell subpopulation analysis. A high-dimensional weighted gene co-expression network analysis (HdWGCNA) identified key molecular features. LASSO regression and Random Survival Forest (RSF) techniques were used to create and validate a prognostic model. Survival analysis, immune microenvironment assessment, and drug sensitivity analysis were conducted. RESULTS Sixteen cell clusters and nine distinct cell types were identified, with T cells showing significant roles in cell communication. The NMF analysis of CD8 +T cells revealed five copper metabolism-related subtypes. The prognostic model based on nine CMRGs indicated significant survival differences between high- and low-risk groups. High-risk patients showed shorter survival times, increased immune cell infiltration, and altered immune responses. Drug sensitivity analysis suggested higher efficacy of certain drugs in high-CMRG patients.
Collapse
Affiliation(s)
- Yi Chen
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Yunmei Liao
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Lang Huang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Zhibin Luo
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China.
| |
Collapse
|
42
|
Huang X, Lian M, Li C. Copper homeostasis and cuproptosis in gynecological cancers. Front Cell Dev Biol 2024; 12:1459183. [PMID: 39386020 PMCID: PMC11461353 DOI: 10.3389/fcell.2024.1459183] [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: 07/03/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
Copper (Cu) is an essential trace element involved in a variety of biological processes, such as antioxidant defense, mitochondrial respiration, and bio-compound synthesis. In recent years, a novel theory called cuproptosis has emerged to explain how Cu induces programmed cell death. Cu targets lipoylated enzymes in the tricarboxylic acid cycle and subsequently triggers the oligomerization of lipoylated dihydrolipoamide S-acetyltransferase, leading to the loss of Fe-S clusters and induction of heat shock protein 70. Gynecological malignancies including cervical cancer, ovarian cancer and uterine corpus endometrial carcinoma significantly impact women's quality of life and even pose a threat to their lives. Excessive Cu can promote cancer progression by enhancing tumor growth, proliferation, angiogenesis and metastasis through multiple signaling pathways. However, there are few studies investigating gynecological cancers in relation to cuproptosis. Therefore, this review discusses Cu homeostasis and cuproptosis while exploring the potential use of cuproptosis for prognosis prediction as well as its implications in the progression and treatment of gynecological cancers. Additionally, we explore the application of Cu ionophore therapy in treating gynecological malignancies.
Collapse
Affiliation(s)
- Xiaodi Huang
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Obstetrics and Gynecology, Shenzhen PKU-HKUST Medical Center, Shenzhen, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, China
| | - Mengyi Lian
- Department of Obstetrics and Gynecology, Longquan People’s Hospital, Lishui, China
| | - Changzhong Li
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Obstetrics and Gynecology, Shenzhen PKU-HKUST Medical Center, Shenzhen, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, China
| |
Collapse
|
43
|
Zeidler-Erdely PC, Kodali V, Falcone LM, Mercer R, Leonard SS, Stefaniak AB, Grose L, Salmen R, Trainor-DeArmitt T, Battelli LA, McKinney W, Stone S, Meighan TG, Betler E, Friend S, Hobbie KR, Service S, Kashon M, Antonini JM, Erdely A. Absence of lung tumor promotion with reduced tumor size in mice after inhalation of copper welding fumes. Carcinogenesis 2024; 45:630-641. [PMID: 39046922 DOI: 10.1093/carcin/bgae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
Welding fumes are a Group 1 (carcinogenic to humans) carcinogen as classified by the International Agency for Research on Cancer. The process of welding creates inhalable fumes rich in iron (Fe) that may also contain known carcinogenic metals such as chromium (Cr) and nickel (Ni). Epidemiological evidence has shown that both mild steel (Fe-rich) and stainless steel (Fe-rich + Cr + Ni) welding fume exposure increases lung cancer risk, and experimental animal data support these findings. Copper-nickel (CuNi) welding processes have not been investigated in the context of lung cancer. Cu is intriguing, however, given the role of Cu in carcinogenesis and cancer therapeutics. This study examines the potential for a CuNi fume to induce mechanistic key characteristics of carcinogenesis in vitro and to promote lung tumorigenesis, using a two-stage mouse bioassay, in vivo. Male A/J mice, initiated with 3-methylcholanthrene (MCA; 10 µg/g), were exposed to CuNi fumes or air by whole-body inhalation for 9 weeks (low deposition-LD and high deposition-HD) and then sacrificed at 30 weeks. In BEAS-2B cells, the CuNi fume-induced micronuclei and caused DNA damage as measured by γ-H2AX. The fume exhibited high reactivity and a dose-response in cytotoxicity and oxidative stress. In vivo, MCA/CuNi HD and LD significantly decreased lung tumor size and adenomas. MCA/CuNi HD exposure significantly decreased gross-evaluated tumor number. In summary, the CuNi fume in vitro exhibited characteristics of a carcinogen, but in vivo, the exposure resulted in smaller tumors, fewer adenomas, less hyperplasia severity, and with HD exposure, less overall lung lesions/tumors.
Collapse
Affiliation(s)
- Patti C Zeidler-Erdely
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Vamsi Kodali
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Lauryn M Falcone
- Department of Dermatology, University of Pittsburgh Medical Center, 3708 Fifth Avenue Suite 500.68, Pittsburgh, PA 15213, United States
| | - Robert Mercer
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Stephen S Leonard
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Aleksandr B Stefaniak
- Respiratory Health Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Lindsay Grose
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Rebecca Salmen
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Taylor Trainor-DeArmitt
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Lori A Battelli
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Walter McKinney
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Samuel Stone
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Terence G Meighan
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Ella Betler
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Sherri Friend
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Kristen R Hobbie
- Pathology Department, Inotiv, P.O. Box 13501, Research Triangle Park, NC 27709, United States
| | - Samantha Service
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Michael Kashon
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - James M Antonini
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| | - Aaron Erdely
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1000 Frederick Lane, Morgantown, WV 26508, United States
| |
Collapse
|
44
|
Tiwari L, Leach C, Williams A, Lighter B, Heiden Z, Roll MF, Moberly JG, Cornell KA, Waynant KV. Binding Mechanisms and Therapeutic Activity of Heterocyclic Substituted Arylazothioformamide Ligands and Their Cu(I) Coordination Complexes. ACS OMEGA 2024; 9:37141-37154. [PMID: 39246472 PMCID: PMC11375723 DOI: 10.1021/acsomega.4c04216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/02/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024]
Abstract
Finding new sources of biologically active compounds for anticancer or antimicrobial therapies remains an active area of research. Azothioformamides (ATFs) with a 1,3 N=N-C=S heterodiene backbone are a new class of biologically active compounds that chelate metals (e.g., Cu) forming stable ATF metal coordination complexes. In this study, ATF ligands were prepared with pyrrolidine, piperidine, N-methylpiperazine, and morpholine substituents on the formamide as to add more heterocyclic drug-like character for biological studies. Formamide derivatives were then complexed with various Cu(I) salts to form coordination complexes. Cu(I) salts were selected as to create potential bioactive compounds with less toxicity. Binding association constants of each Cu(I) salt to ATF ligands were extrapolated from UV-vis titration studies and were corroborated with DFT calculations using a hybrid functional B3LYP method. It was observed that the smaller pyrrolidine functionalized ATFs bound to the Cu(I) salts had stronger binding than any of the larger six-membered-ring heterocycles with association values in the 104 - 105 M-1 range. The ATF-Cu(I) salt coordination complexes were then evaluated for antimicrobial activity against two bacteria (Staphylococcus aureus, Escherichia coli), one yeast (Candida albicans), four human cancer lines (A-549, K-562, HT-1080, MDA-MB-231), and two normal human lines (MRC-5, HFF). The ATF ligands themselves were inactive against all microbes and most human lines except K-562 cells, which were sensitive to three of the four ligands (IC50's = 7.0-25.5 μM). Most ATF-Cu(I) complexes showed low to medium micromolar activity against Candida albicans (IC50's 2.6-24.8 μM) and Staphylococcus aureus (IC50's = 3.4-37.7 μM), with increasing activity corresponding to complexes with higher binding association constants. The antiproliferative properties of ATF-Cu(I) metal salt complexes against mammalian cells were mixed, with low to medium micromolar activity across all cell lines. Notably, several ATF-Cu(I) salt coordination complexes showed submicromolar activity against the HT-1080 fibrosarcoma line (0.52-0.69 μM). The results demonstrate promising activity of ATF-Cu(I) complexes, particularly with pyrrolidine as the formamide component. These studies suggest that the stronger binding association values correlate to higher levels of biological activity.
Collapse
Affiliation(s)
- Laxmi Tiwari
- Department
of Chemistry, University of Idaho, Moscow, Idaho 83844, United States
| | - Caleb Leach
- Department
of Chemistry and Biochemistry, Boise State
University, Boise, Idaho 83725, United States
| | - Ashley Williams
- Department
of Chemistry and Biochemistry, Boise State
University, Boise, Idaho 83725, United States
| | - Brandon Lighter
- Department
of Chemistry and Biochemistry, Boise State
University, Boise, Idaho 83725, United States
| | - Zachariah Heiden
- Department
of Chemistry, Washington State University, Pullman, Washington 99164, United States
| | - Mark F. Roll
- Department
of Mechanical Engineering, University of
Idaho, Moscow, Idaho 83844, United States
| | - James G. Moberly
- Department
of Chemical and Biological Engineering, University of Idaho, Moscow, Idaho 83844, United States
| | - Kenneth A. Cornell
- Department
of Chemistry and Biochemistry, Boise State
University, Boise, Idaho 83725, United States
| | | |
Collapse
|
45
|
Sabolović J. Bis(amino acidato)copper(II) compounds in blood plasma: a review of computed structural properties and amino acid affinities for Cu 2+ informing further pharmacological research. Arh Hig Rada Toksikol 2024; 75:159-171. [PMID: 39369326 PMCID: PMC11456223 DOI: 10.2478/aiht-2024-75-3871] [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: 06/01/2024] [Revised: 06/01/2024] [Accepted: 06/01/2024] [Indexed: 10/07/2024] Open
Abstract
Neutral bis(amino acidato)copper(II) [Cu(aa)2] coordination compounds are the physiological species of copper(II) amino acid compounds in blood plasma taking the form of bis(l-histidinato)copper(II) and mixed ternary copper(II)-l-histidine complexes, preferably with l-glutamine, l-threonine, l-asparagine, and l-cysteine. These amino acids have three functional groups that can bind metal ions: the common α-amino and carboxylate groups and a side-chain polar group. In Cu(aa)2, two coordinating groups per amino acid bind to copper(II) in-plane, while the third group can bind apically, which yields many possibilities for axial and planar bonds, that is, for bidentate and tridentate binding. So far, the experimental studies of physiological Cu(aa)2 compounds in solutions have not specified their complete geometries. This paper provides a brief review of my group's research on structural properties of physiological Cu(aa)2 calculated using the density functional theory (DFT) to locate low-energy conformers that can coexist in aqueous solutions. These DFT investigations have revealed high conformational flexibility of ternary Cu(aa)2 compounds for tridentate or bidentate chelation, which may explain copper(II) exchange reactions in the plasma and inform the development of small multifunctional copper(II)-binding drugs with several possible copper(II)-binding groups. Furthermore, our prediction of metal ion affinities for Cu2+ binding with amino-acid ligands in low-energy conformers with different coordination modes of five physiological Cu(aa)2 in aqueous solution supports the findings of their abundance in human plasma obtained with chemical speciation modelling.
Collapse
Affiliation(s)
- Jasmina Sabolović
- Institute for Medical Research and Occupational Health, Division of Occupational and Environmental Health, Zagreb, Croatia
| |
Collapse
|
46
|
Joshi A, Mandal R. Review Article on Molecular Basis of Zinc and Copper Interactions in Cancer Physiology. Biol Trace Elem Res 2024:10.1007/s12011-024-04356-5. [PMID: 39215955 DOI: 10.1007/s12011-024-04356-5] [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: 06/19/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Various clinical manifestations associated with measurable abnormalities of Zn and Cu in serum and tissue were determined in Cancer-Patients (CP), and therefore, these two metals are drawing more and more attention presently than ever before. Cancer is a disease of uncontrolled-abnormal-cell-division with invasion-potential which was exhibited to occur due to dys-regulation/dys-homeostasis of fundamental-biological-pathways (FBP) including antioxidant-enzyme-defense-system, anti-inflammatory and immune-systems, and DNA-damage-repair-system in the human-body resulting in generation of chronic-oxidative-stress induced DNA-damage and gene-mutations, inflammation and compromised immune-system, tumor-induced increased angiogenesis, and inhibition of apoptosis processes. Zn and Cu were recognized to be the most crucial components of FBP and imbalance in Zn/Cu ratios in CP asserted to generate chronic toxicity in human body through various mechanisms including increased chronic oxidative stress linked compromised DNA integrity and gene mutations due to malfunctioning of DNA damage repair enzymes; increased angiogenesis process due to Zn- and Cu-binding proteins metallothionein and ceruloplasmin-induced enhanced expression of tumor growth factors; and elevation in inflammatory response which was further shown to down/upregulate gene expression of multiple Zn transporter proteins leading to dys-homeostasis of intracellular Zn concentrations, and it was determined to disturb the equilibrium between cell growth and division, proliferation, differentiation, and apoptosis processes which lead to cancer progression. Moreover, Zn was reported to affect matrix metalloproteinase activity and influence immune system cells to respond differently to different cytokines and enhance immune-suppressive effects accelerating the angiogenesis, invasion, and metastasis potential in cancer. Further, the most significant use of serum Cu/Zn ratio was recommended in clinical diagnosis, prognosis, tumor stage, patient survival, and cancer follow-up studies which need further investigations to elucidate and explore their roles in cancer physiology for clinical perspective.
Collapse
Affiliation(s)
- Amit Joshi
- PG Department of Biotechnology and Microbial Biotechnology, Sri Guru Gobind Singh College, Chandigarh, UT, India
| | - Reshu Mandal
- PG Department of Zoology, Sri Guru Gobind Singh College, Chandigarh, UT, India.
| |
Collapse
|
47
|
Salloom RJ, Ahmad IM, Sahtout DZ, Baine MJ, Abdalla MY. Heme Oxygenase-1 and Prostate Cancer: Function, Regulation, and Implication in Cancer Therapy. Int J Mol Sci 2024; 25:9195. [PMID: 39273143 PMCID: PMC11394971 DOI: 10.3390/ijms25179195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Prostate cancer (PC) is a significant cause of mortality in men worldwide, hence the need for a comprehensive understanding of the molecular mechanisms underlying its progression and resistance to treatment. Heme oxygenase-1 (HO-1), an inducible enzyme involved in heme catabolism, has emerged as a critical player in cancer biology, including PC. This review explores the multifaceted role of HO-1 in PC, encompassing its function, regulation, and implications in cancer therapy. HO-1 influences cell proliferation, anti-apoptotic pathways, angiogenesis, and the tumor microenvironment, thereby influencing tumor growth and metastasis. HO-1 has also been associated with therapy resistance, affecting response to standard treatments. Moreover, HO-1 plays a significant role in immune modulation, affecting the tumor immune microenvironment and potentially influencing therapy outcomes. Understanding the intricate balance of HO-1 in PC is vital for developing effective therapeutic strategies. This review further explores the potential of targeting HO-1 as a therapeutic approach, highlighting challenges and opportunities. Additionally, clinical implications are discussed, focusing on the prognostic value of HO-1 expression and the development of novel combined therapies to augment PC sensitivity to standard treatment strategies. Ultimately, unraveling the complexities of HO-1 in PC biology will provide critical insights into personalized treatment approaches for PC patients.
Collapse
Affiliation(s)
- Ramia J. Salloom
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.J.S.); (D.Z.S.)
| | - Iman M. Ahmad
- Department of Clinical, Diagnostic, and Therapeutic Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Dania Z. Sahtout
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.J.S.); (D.Z.S.)
| | - Michael J. Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Maher Y. Abdalla
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.J.S.); (D.Z.S.)
| |
Collapse
|
48
|
Lai Y, Gao FF, Ge RT, Liu R, Ma S, Liu X. Metal ions overloading and cell death. Cell Biol Toxicol 2024; 40:72. [PMID: 39162885 PMCID: PMC11335907 DOI: 10.1007/s10565-024-09910-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
Cell death maintains cell morphology and homeostasis during development by removing damaged or obsolete cells. The concentration of metal ions whithin cells is regulated by various intracellular transporters and repositories to maintain dynamic balance. External or internal stimuli might increase the concentration of metal ions, which results in ions overloading. Abnormal accumulation of large amounts of metal ions can lead to disruption of various signaling in the cell, which in turn can produce toxic effects and lead to the occurrence of different types of cell deaths. In order to further study the occurrence and development of metal ions overloading induced cell death, this paper reviewed the regulation of Ca2+, Fe3+, Cu2+ and Zn2+ metal ions, and the internal mechanism of cell death induced by overloading. Furthermore, we found that different metal ions possess a synergistic and competitive relationship in the regulation of cell death. And the enhanced level of oxidative stress was present in all the processes of cell death due to metal ions overloading, which possibly due to the combination of factors. Therefore, this review offers a theoretical foundation for the investigation of the toxic effects of metal ions, and presents innovative insights for targeted regulation and therapeutic intervention. HIGHLIGHTS: • Metal ions overloading disrupts homeostasis, which in turn affects the regulation of cell death. • Metal ions overloading can cause cell death via reactive oxygen species (ROS). • Different metal ions have synergistic and competitive relationships for regulating cell death.
Collapse
Affiliation(s)
- Yun Lai
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Fen Fen Gao
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Ruo Ting Ge
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Rui Liu
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Shumei Ma
- School of Public Health, Wenzhou Medical University, Wenzhou, China.
| | - Xiaodong Liu
- School of Public Health, Wenzhou Medical University, Wenzhou, China.
- South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou Medical University, Wenzhou, China.
- Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
49
|
Ross MO, Xie Y, Owyang RC, Ye C, Zbihley ONP, Lyu R, Wu T, Wang P, Karginova O, Olopade OI, Zhao M, He C. PTPN2 copper-sensing relays copper level fluctuations into EGFR/CREB activation and associated CTR1 transcriptional repression. Nat Commun 2024; 15:6947. [PMID: 39138174 PMCID: PMC11322707 DOI: 10.1038/s41467-024-50524-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Fluxes in human copper levels recently garnered attention for roles in cellular signaling, including affecting levels of the signaling molecule cyclic adenosine monophosphate. We herein apply an unbiased temporal evaluation of the signaling and whole genome transcriptional activities modulated by copper level fluctuations to identify potential copper sensor proteins responsible for driving these activities. We find that fluctuations in physiologically relevant copper levels modulate EGFR signal transduction and activation of the transcription factor CREB. Both intracellular and extracellular assays support Cu1+ inhibition of the EGFR phosphatase PTPN2 (and potentially PTPN1)-via ligation to the PTPN2 active site cysteine side chain-as the underlying mechanism. We additionally show i) copper supplementation drives weak transcriptional repression of the copper importer CTR1 and ii) CREB activity is inversely correlated with CTR1 expression. In summary, our study reveals PTPN2 as a physiological copper sensor and defines a regulatory mechanism linking feedback control of copper stimulated EGFR/CREB signaling and CTR1 expression.
Collapse
Affiliation(s)
- Matthew O Ross
- Department of Chemistry, University of Chicago, Chicago, IL, USA.
| | - Yuan Xie
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Ryan C Owyang
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Chang Ye
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Olivia N P Zbihley
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Ruitu Lyu
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Tong Wu
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Pingluan Wang
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Olga Karginova
- Department of Medicine, Center for Clinical Cancer Genetics and Global Health, University of Chicago, Chicago, IL, USA
| | - Olufunmilayo I Olopade
- Department of Medicine, Center for Clinical Cancer Genetics and Global Health, University of Chicago, Chicago, IL, USA
| | - Minglei Zhao
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Chuan He
- Department of Chemistry, University of Chicago, Chicago, IL, USA.
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA.
- Howard Hughes Medical Institute, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
50
|
Satapathi D, Das M, Das UK, Laha S, Kundu P, Choudhuri I, Bhattacharya N, Samanta BC, Chattopadhyay N, Maity T. Experimental and molecular modelling demonstration of effective DNA and protein binding as well as anticancer potential of two mononuclear Cu(II) and Co(II) complexes with isothiocyanate and azide as anionic residues. Int J Biol Macromol 2024; 275:133716. [PMID: 38977049 DOI: 10.1016/j.ijbiomac.2024.133716] [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: 03/27/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
In the present study, one mononuclear Cu(II) [CuL(SCN)] (1) and one mononuclear Co(II) [CoLN3] (2) complexes, with a Schiff base ligand (HL) formed by condensation of 2-picolylamine and salicylaldehyde, have been successfully developed and structurally characterized. The square planer geometry of both complexes is fulfilled by the coordination of one deprotonated ligand and one ancillary ligand SCN-(1) or N3-(2) to the metal centre. Binding affinities of both complexes with deoxyribonucleic acid (DNA) and human serum albumin (HSA) are investigated using several biophysical and spectroscopic techniques. High values of the macromolecule-complex binding constants and other results confirm the effectiveness of both complexes towards binding with DNA and HSA. The determined values of the thermodynamic parameters support spontaneous interactions of both complexes with HSA, while fluorescence displacement and DNA melting studies establish groove-binding interactions with DNA for both complexes 1 and 2. The molecular modelling study validates the experimental findings. Both complexes are subjected to an MTT test establishing the anticancer property of complex 1 with lower risk to normal cells, confirmed by the IC50 values of the complex for HeLa cancer cells and HEK normal cells. Finally, a nuclear staining analysis reveals that the complexes have caused apoptotic cell death.
Collapse
Affiliation(s)
- Dibyendu Satapathi
- Department of Chemistry, Prabhat Kumar College, Purba Medinipur, Contai, West Bengal 721404, India
| | - Manik Das
- Department of Chemistry, Prabhat Kumar College, Purba Medinipur, Contai, West Bengal 721404, India
| | - Uttam Kumar Das
- Department of Chemistry, School of Physical Science, Mahatma Gandhi Central University, Bihar, India
| | - Soumik Laha
- Indian Institute of Chemical Biology, Jadavpur, Kolkata, West Bengal, India
| | - Pronab Kundu
- Department of Chemistry, Presidency University, Yelahanka, Bengaluru 560064, India
| | - Indranil Choudhuri
- Department of Biotechnology, Panskura Banamali College, Panskura, West Bengal, India
| | - Nandan Bhattacharya
- Department of Biotechnology, Panskura Banamali College, Panskura, West Bengal, India
| | | | | | - Tithi Maity
- Department of Chemistry, Prabhat Kumar College, Purba Medinipur, Contai, West Bengal 721404, India.
| |
Collapse
|