1
|
Feng D, Kang X, Wang H, He Z, Xu H, Li Y, Fan A, Xu H, Zhang Y, Song J, Hou J, Qi J, Zhang W. Photochemical bomb: Precision nuclear targeting to activate cGAS-STING pathway for enhanced bladder cancer immunotherapy. Biomaterials 2025; 318:123126. [PMID: 39884129 DOI: 10.1016/j.biomaterials.2025.123126] [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: 10/21/2024] [Revised: 12/31/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Activating the cGAS-STING pathway presents a promising strategy to enhance the innate immunity and combat the immunosuppressive tumor microenvironment. One key mechanism for triggering this pathway involves the release of damaged DNA fragments caused by nuclear DNA damage. However, conventional cGAS-STING agonists often suffer from limited nucleus-targeting efficiency and potential biotoxicity. In this study, we develop a novel nucleus-targeting theranostic nanoplatform designed to synergistically activate the cGAS-STING pathway through the combination of photodynamic therapy (PDT) and cisplatin chemotherapy for orthotopic bladder cancer treatment. The nanoplatform integrates a new high-performance type-I photosensitizer with near-infrared-II emission, a TATSA peptide for enhanced nuclear targeting, and a biosafe platinum (IV) cisplatin prodrug. Upon NIR laser irradiation, the nanoagent delivers synergistic nucleus-targeted PDT and chemotherapy, causing substantial DNA damage and the release of double-stranded DNA, which subsequently activates the cGAS-STING pathway and triggers potent immunomodulation. This activation promotes dendritic cells maturation, enhances cytotoxic T infiltration, and facilitates the formation of memory T cells, leading to immune microenvironment remodeling, and long-lasting immune memory, thus effectively inhibiting orthotopic bladder tumors and reducing the risk of metastasis. These findings highlight the substantial potential of this strategy to overcome the limitations of current immunotherapies by leveraging nucleus-targeted PDT to activate the cGAS-STING pathway for cancer treatment.
Collapse
Affiliation(s)
- Dexiang Feng
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xiaoying Kang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - He Wang
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Zhangxin He
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Haodong Xu
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yue Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Aohua Fan
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, 130012, China
| | - Hongbo Xu
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China
| | - Yuan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jianwen Song
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jianquan Hou
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Ji Qi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Weijie Zhang
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215000, China; Department of Urology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
2
|
Wu H, Ning Y, Sun Z, Ji J, Lu M, Jiao X, Xu X, Ding X, Cheng X, Yu X. Both carvedilol and cimetidine alleviate cisplatin-induced nephrotoxicity via downregulating OCT2. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167754. [PMID: 40044066 DOI: 10.1016/j.bbadis.2025.167754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/01/2025] [Accepted: 02/25/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Cisplatin is a common chemotherapy agent for solid tumors but severe nephrotoxicity limits its application, with no effective pharmacological treatments. Organic cation transporter 2 (OCT2) is involved in cisplatin uptake in kidneys. This study aimed to find drugs with promising clinical applications that could prevent cisplatin-induced acute kidney injury (Cis-AKI) by inhibiting OCT2. METHODS The mRNA level of OCT2 was examined in human induced pluripotent stem cells (iPSCs) from Cis-AKI patients and paired non-AKI patients. The association between OCT2 and Cis-AKI was investigated by HEK293FT cells and kidney organoids. We screened potential compounds exhibiting protective effects against Cis-AKI in US Food and Drug Administration-approved drugs through virtual screening and activity screening. Subsequently, we determined the effects of these compounds on OCT2 expression, cisplatin uptake, and apoptosis in cells, kidney organoids and mice. A549 and HeLa cells were adopted to observe the influence of drugs on the anti-tumor function of cisplatin. RESULTS Compared to non-AKI patients, the OCT2 mRNA levels of iPSCs from Cis-AKI patients were elevated. OCT2 exhibits similar expression patterns in kidney organoids and human kidney tissues. Furthermore, the overexpression of OCT2 in kidney organoids and HEK293FT cells exacerbated the injury caused by cisplatin. Carvedilol and cimetidine were identified as potent OCT2 inhibitors by drug screening. Further analysis revealed that the pretreatment of carvedilol or cimetidine downregulated OCT2, reduced cisplatin uptake, and alleviated cisplatin-induced apoptosis, but the combination of the two drugs didn't further improve these outcomes. Additionally, carvedilol and cimetidine didn't compromise the cisplatin-induced cell death in A549 and HeLa cells. CONCLUSION Our study confirmed that carvedilol and cimetidine exert protective effects against Cis-AKI by inhibiting OCT2, without altering the anti-tumor effects of cisplatin.
Collapse
Affiliation(s)
- Huan Wu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Yichun Ning
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China; Shanghai Medical Center for Kidney, Shanghai, China; Shanghai Institute of Kidney and Dialysis, Shanghai, China; Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Zhaoxing Sun
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Ji Ji
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Min Lu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Xiaoyan Jiao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China; Shanghai Medical Center for Kidney, Shanghai, China; Shanghai Institute of Kidney and Dialysis, Shanghai, China; Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xiaoliang Xu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China; Shanghai Medical Center for Kidney, Shanghai, China; Shanghai Institute of Kidney and Dialysis, Shanghai, China; Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xin Cheng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Xiaofang Yu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China; Shanghai Medical Center for Kidney, Shanghai, China; Shanghai Institute of Kidney and Dialysis, Shanghai, China; Hemodialysis Quality Control Center of Shanghai, Shanghai, China.
| |
Collapse
|
3
|
Petrova Z, Mocanu T, Spasov R, Hanganu A, Marinescu G, Culita DC, Alexandrova R. Antitumor activity of ruthenium(III) complexes with [N 2O 2]-tetradentate Schiff base ligands. J Inorg Biochem 2025; 266:112853. [PMID: 39946799 DOI: 10.1016/j.jinorgbio.2025.112853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 03/01/2025]
Abstract
In this article, the antitumor and antiproliferative activity of three Ru(III) complexes, [RuIII(Salen)(PPh3)Cl] (RuSalen), [RuIII(Salphen)(PPh3)Cl] (RuSalphen), and [RuIII(Salpn)(PPh3)Cl] (RuSalpn) (H2Salen, H2Salphen and H2Salpn are the Schiff bases obtained by the condensation between salicylaldehyde and ethylenediamine, 1,2-phenylenediamine, and 1,3-diaminopropanne, respectively) and their precursor, [RuII(PPh3)3Cl2], were investigated against laboratory-cultured tumor cell lines: HT29 (human colorectal carcinoma), Saos-2 (human osteogenic sarcoma), HeLa (human cervical carcinoma), RST (rat transplantable sarcoma), and the non-tumor cell line Lep3 (embryonal human fibroblasts). It was found that all the cancer cell lines investigated were effectively dose-dependently inhibited in their growth by the Ru(III) complexes, while the non-tumor cell line Lep3 was the least affected by their cytotoxic effect. The Annexin V assay revealed that the Ru(III) complexes determined the occurrence of apoptosis in all cell lines tested, in a dose-dependent manner. RuSalpn exhibited the strongest ability to reduce tumor cell survival and proliferation, with efficacy that is either superior to or comparable to that of well-established clinical oncology agents such as cisplatin, oxaliplatin, epirubicin, and paclitaxel. The experiments revealed a cell-specific response, with varying degrees of sensitivity to the tested substances across different cell lines. RuSalpn demonstrated the strongest cytotoxic effect in the HT29 cell line, while RuSalen, RuSalphen showed the highest activity against RST cells. It was found that RuSalphen (≥7.0 μM) significantly inhibited cell migratory activity in the HT29 cell line, while in the RST cell line, RuSalen (≥37.6 μM), RuSalphen (≥14.0 μM), and RuSalpn (≥36.8 μM) demonstrated a strong inhibitory effect on cell migration.
Collapse
Affiliation(s)
- Zdravka Petrova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Teodora Mocanu
- Ilie Murgulescu Institute of Physical Chemistry, Romanian Academy, 202 Splaiul Independentei, Bucharest, Romania
| | - Rossen Spasov
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria; Faculty of Medicine, Sofia University "St. Kliment Ohridski", Sofia, Bulgaria
| | - Anamaria Hanganu
- Faculty of Chemistry, University of Bucharest, Regina Elisabeta Blvd. 4-12, Bucharest, Romania; C.D. Nenitzescu" Institute of Organic and Supramolecular Chemistry of the Romanian Academy, 202B Splaiul Independentei, Bucharest, Romania
| | - Gabriela Marinescu
- Ilie Murgulescu Institute of Physical Chemistry, Romanian Academy, 202 Splaiul Independentei, Bucharest, Romania
| | - Daniela C Culita
- Ilie Murgulescu Institute of Physical Chemistry, Romanian Academy, 202 Splaiul Independentei, Bucharest, Romania.
| | - Radostina Alexandrova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| |
Collapse
|
4
|
Wu Y, Yang L, Li Z, Chen Q, Hu J. Polyphyllin VII Enhances the Antitumor Activity of Cisplatin in Non-Small Cell Lung Cancer Cells by Inducing Ferroptosis and Enhancing Apoptosis. J Biochem Mol Toxicol 2025; 39:e70186. [PMID: 40165507 DOI: 10.1002/jbt.70186] [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: 10/17/2024] [Revised: 01/06/2025] [Accepted: 02/08/2025] [Indexed: 04/02/2025]
Abstract
Cisplatin (DDP) resistance in non-small cell lung cancer (NSCLC) is a common cause of treatment failure and a significant contributor to increased mortality. To tackle this issue, the integration of traditional Chinese medicine with chemotherapy has been proposed as a promising approach. The potential synergistic effect of combining polyphyllin VII (PPVII) and DDP in overcoming DDP resistance in NSCLC cells has not been thoroughly investigated yet. In this study, H1299 cells were exposed to gradient concentrations of PPVII and DDP to determine their 50% inhibitory concentration values, and the most effective concentration was applied in subsequent experiments. The combination of PPVII and DDP was evaluated for its effects on H1299 cell proliferation, apoptosis, viability, and the expression of proteins linked to apoptosis and ferroptosis. To further elucidate the underlying mechanisms, the impact of the combination on DNA damage in H1299 cells was also examined. Our results demonstrated that PPVII significantly potentiated the antitumor effects of DDP in H1299 cells in a dose-dependent manner (p < 0.05). Furthermore, PPVII was observed to work synergistically with DDP to suppress proliferation and promote apoptosis in H1299 cells (p < 0.05). Western blotting analysis proved that the combination treatment upregulated proapoptotic proteins (B-cell lymphoma 2-associated X protein, cleaved-caspase 3 and cleaved-PARP), downregulated antiapoptotic protein (Bcl-2), and promoted ferroptosis-associated proteins (long-chain acyl-coenzyme A synthase 4 and NADPH oxidase 4) as well as DNA damage-associated protein (γH2AX) (p < 0.05). Overall, the combination of PPVII and DDP significantly enhanced antitumor activity in H1299 cells through the modulation of DNA damage and ferroptosis, suggesting its potential as an effective therapeutic approach against DDP-resistant NSCLC.
Collapse
Affiliation(s)
- Yuanzhou Wu
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Liang Yang
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zizhao Li
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qunqing Chen
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jia Hu
- Department Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Mboni-Johnston IM, Hartmann S, Kroll C, Berndt C, Adjaye J, Schupp N. Impact of nephrotoxins and oxidants on survival and transport function of hiPSC-derived renal proximal tubular cells. Arch Toxicol 2025:10.1007/s00204-025-04015-1. [PMID: 40119912 DOI: 10.1007/s00204-025-04015-1] [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] [Accepted: 02/27/2025] [Indexed: 03/25/2025]
Abstract
Due to their role in excretion, renal proximal tubular cells are susceptible to damage by toxic metabolites and xenobiotics. The regenerative capacity of the kidney allows for the replacement of damaged cells, a process involving differentiation programs. However, kidney function tends to decline, suggesting that the replacement cells may not achieve full functionality. To understand possible causes of this decline, we investigated effects of nephrotoxins and oxidants on the differentiation of induced pluripotent stem cells (iPSC) into proximal tubular epithelial-like cells (PTELC). Proliferation, apoptosis, senescence, and expression of oxidative defense genes were analyzed in iPSC, differentiating and differentiated cells treated with cisplatin (CisPt, up to 45 µM), cyclosporin A (CycA, up to 12 µM), and the oxidants menadione (Mena, up to 50 µM) and tert-butylhydroquinone (tBHQ, up to 50 µM). We found that differentiating cells were most sensitive to oxidants and showed increased sensitivity to CisPt, whereas all differentiation stages showed similar sensitivity to CycA. Both oxidative stress and CisPt triggered apoptosis in all differentiation stages, whereas CycA mainly induced senescence. Treatment during differentiation resulted in long-term effects on gene expression in differentiated cells. While oxidants had no effect on transport function of differentiated cells, CisPt and CycA impaired albumin uptake. Our data suggest a substantial sensitivity of differentiating cells to nephrotoxins and oxidants, an aspect that could potentially interfere with regenerative processes.
Collapse
Affiliation(s)
- Isaac Musong Mboni-Johnston
- Institute of Toxicology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Sören Hartmann
- Institute of Toxicology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Christian Kroll
- Department of Neurology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, University Hospital Düsseldorf, University of Düsseldorf, 40225, Düsseldorf, Germany
- Zayed Centre for Research into Rare Diseases in Children (ZCR), EGA Institute for Women'S Health, University College London (UCL), 20 Guilford Street, London, WC1N 1DZ, UK
| | - Nicole Schupp
- Institute of Toxicology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
6
|
Jin Y, Xue B, Zhou X. Protein Biomarkers of DNA Damage in Yeast Cells for Genotoxicity Screening. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:250-258. [PMID: 40144325 PMCID: PMC11934195 DOI: 10.1021/envhealth.4c00160] [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: 08/22/2024] [Revised: 10/27/2024] [Accepted: 11/01/2024] [Indexed: 03/28/2025]
Abstract
Providing an unbiased and comprehensive view of the DNA damage response in cells is critical in genotoxicity screening to identify substances that cause diverse types of DNA damage. Considering that S. cerevisiae is one of the most well-characterized model organisms in molecular and cellular biology, we created a map of the DNA damage response network containing the reported signaling pathways in yeast cells programmed to constitutively respond to DNA damage. A collection of GFP-fused S. cerevisiae yeast strains treated with typical genotoxic agents illuminated the cellular response to DNA damage, thereby identifying 15 protein biomarkers encompassing all eight documented DNA damage response pathways. Three statistical and one deep learning models were proposed to interpret the quantitative molecular toxicity end point, i.e. protein effect level index (PELI), by introducing weights of 15 biomarkers in genotoxicity assessment. The method based on standard deviation exhibited the best performance, with an R 2 of 0.916 compared to the SOS/umu test and an R 2 of 0.989 compared to the comet assay. The GFP-fused yeast-based proteomic assay has minute-level resolution of pathway activation data. It provides a concise alternative for fast, efficient, and mechanistic genotoxicity screening for various environmental and health applications.
Collapse
Affiliation(s)
- Yushi Jin
- Center for Sensor Technology of Environment
and Health, School of Environment, Tsinghua
University, Beijing 100084, China
| | - Boyuan Xue
- Center for Sensor Technology of Environment
and Health, School of Environment, Tsinghua
University, Beijing 100084, China
| | - Xiaohong Zhou
- Center for Sensor Technology of Environment
and Health, School of Environment, Tsinghua
University, Beijing 100084, China
| |
Collapse
|
7
|
Shi J, Liu W, Song A, Sanni T, Van Deusen A, Zunder ER, Deppmann CD. Extrinsic Apoptosis and Necroptosis in Telencephalic Development: A Single-Cell Mass Cytometry Study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.01.640843. [PMID: 40093055 PMCID: PMC11908208 DOI: 10.1101/2025.03.01.640843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Regulated cell death is integral to sculpting the developing brain, yet the relative contributions of extrinsic apoptosis and necroptosis remain unclear. Here, we leverage single-cell mass cytometry (CyTOF) to characterize the cellular landscape of the mouse telencephalon in wild-type (WT), RIPK3 knockout (RIPK3 KO), and RIPK3/Caspase-8 double knockout (DKO) mice. Strikingly, combined deletion of RIPK3 and Caspase-8 leads to a 12.6% increase in total cell count, challenging the prevailing notion that intrinsic apoptosis exclusively governs developmental cell elimination. Detailed subpopulation analysis reveals that DKO mice display selective enrichment of Tbr2⁺ intermediate progenitors and endothelial cells, underscoring distinct, cell type-specific roles for extrinsic apoptotic and necroptotic pathways. These findings provide a revised framework for understanding the coordinated regulation of cell number during telencephalic development and suggest potential mechanistic links to neurodevelopmental disorders characterized by aberrant cell death.
Collapse
|
8
|
Wagner AM, Lanier OL, Savk A, Peppas NA. Polybasic nanogels for intracellular co-delivery of paclitaxel and carboplatin: a novel approach to ovarian cancer therapy. RSC PHARMACEUTICS 2025:d4pm00330f. [PMID: 39990011 PMCID: PMC11843545 DOI: 10.1039/d4pm00330f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
Ovarian cancer is one of the leading causes of cancer-related deaths in women, with limited progress in treatments despite decades of research. Common treatment protocols rely on surgical removal of tumors and chemotherapy drugs, such as paclitaxel and carboplatin, which are capable of reaching cancer cells throughout the body. However, the effectiveness of these drugs is often limited due to toxic reactions in patients, nonspecific drug distribution affecting healthy cells, and the development of treatment resistance. In this study, we introduce a polybasic nanogel system composed of poly(diethylaminoethyl methacrylate-co-cyclohexyl methacrylate)-g-poly(ethylene glycol) designed for the targeted co-delivery of paclitaxel and carboplatin directly to ovarian cancer cells. These nanogel systems can respond to the cellular microenvironment to achieve controlled, on-demand drug release, reducing off-target effects and enhancing therapeutic uptake. Additionally, we investigated nanoparticle degradation and controlled drug release as a function of various crosslinkers, including tetraethylene glycol dimethacrylate, bis(2-methacryloyl)oxyethyl disulfide, poly(lactic acid)-b-poly(ethylene glycol)-b-poly(lactic acid)dimethacrylate, and polycaprolactone dimethacrylate. Our results, using OVCAR-3 human ovarian cancer cells, demonstrated that this dual-delivery system outperformed free drugs in inducing cancer cell death, representing a promising advance in the field of nanoparticle-based therapies for ovarian cancer. By loading two chemotherapeutic agents into a single, environmentally responsive particle, this approach shows the potential to overcome common resistance mechanisms and achieve more effective tumor suppression. In summary, by delivering chemotherapy more precisely, it may be possible to enhance therapeutic outcomes while minimizing toxicity and nonspecific drug distribution, ultimately improving patient quality of life.
Collapse
Affiliation(s)
- Angela M Wagner
- McKetta Department of Chemical Engineering, The University of Texas at Austin Austin TX USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin Austin TX USA
| | - Olivia L Lanier
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin Austin TX USA
- Department of Biomedical Engineering, The University of Texas at Austin Austin TX USA
- Department of Chemical and Biological Engineering, University of New Mexico Albuquerque NM USA
- Department of Biomedical Engineering, University of New Mexico Albuquerque NM USA
- Cancer Therapeutics Program, University of New Mexico Comprehensive Cancer Center Albuquerque NM USA
| | - Ani Savk
- McKetta Department of Chemical Engineering, The University of Texas at Austin Austin TX USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin Austin TX USA
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, The University of Texas at Austin Austin TX USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin Austin TX USA
- Department of Biomedical Engineering, The University of Texas at Austin Austin TX USA
- Department of Surgery and Perioperative Care, Dell Medical School, University of Texas at Austin Austin TX USA
- Department of Pediatrics, Dell Medical School, University of Texas at Austin Austin TX USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin Austin TX USA
| |
Collapse
|
9
|
Chen SH, Yu JH, Lin YC, Chang YM, Liu NT, Chen SF. Application of an Integrated Single-Cell and Three-Dimensional Spheroid Culture Platform for Investigating Drug Resistance Heterogeneity and Epithelial-Mesenchymal Transition (EMT) in Lung Cancer Subclones. Int J Mol Sci 2025; 26:1766. [PMID: 40004228 PMCID: PMC11855057 DOI: 10.3390/ijms26041766] [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: 12/21/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Lung cancer is a leading cause of cancer-related mortality worldwide, largely due to its heterogeneity and intrinsic drug resistance. Malignant pleural effusions (MPEs) provide diverse tumor cell populations ideal for studying these complexities. Although chemotherapy and targeted therapies can be initially effective, subpopulations of cancer cells with phenotypic plasticity often survive treatment, eventually developing resistance. Here, we integrated single-cell isolation and three-dimensional (3D) spheroid culture to dissect subclonal heterogeneity and drug responses, aiming to inform precision medicine approaches. Using A549 lung cancer cells, we established a cisplatin-resistant line and isolated three resistant subclones (Holoclone, Meroclone, Paraclone) via single-cell sorting. In 3D spheroids, Docetaxel and Alimta displayed higher IC50 values than in 2D cultures, suggesting that 3D models better reflect clinical dosing. Additionally, MPE-derived Holoclone and Paraclone subclones exhibited distinct sensitivities to Giotrif and Capmatinib, revealing their heterogeneous drug responses. Molecular analyses confirmed elevated ABCB1, ABCG2, cancer stem cell (CSC) markers (OCT4, SOX2, CD44, CD133), and epithelial-mesenchymal transition (EMT) markers (E-cadherin downregulation, increased Vimentin, N-cadherin, Twist) in resistant subclones, correlating with enhanced migration and invasion. This integrated approach clarifies the interplay between heterogeneity, CSC/EMT phenotypes, and drug resistance, providing a valuable tool for predicting therapeutic responses and guiding personalized, combination-based lung cancer treatments.
Collapse
Affiliation(s)
- Shin-Hu Chen
- Department of Dentistry, School of Dentistry, China Medical University, Taichung 40403, Taiwan; (S.-H.C.); (J.-H.Y.)
| | - Jian-Hong Yu
- Department of Dentistry, School of Dentistry, China Medical University, Taichung 40403, Taiwan; (S.-H.C.); (J.-H.Y.)
| | - Yu-Chun Lin
- Department of Pathology, National Defense Medical Center, Tri-Service General Hospital, Taipei 114201, Taiwan; (Y.-C.L.); (N.-T.L.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
- Institute of Pathology and Parasitology, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Yi-Ming Chang
- Institute of Pathology and Parasitology, National Defense Medical Center, Taipei 11490, Taiwan;
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
| | - Nien-Tzu Liu
- Department of Pathology, National Defense Medical Center, Tri-Service General Hospital, Taipei 114201, Taiwan; (Y.-C.L.); (N.-T.L.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
| | - Su-Feng Chen
- Department of Dentistry, School of Dentistry, China Medical University, Taichung 40403, Taiwan; (S.-H.C.); (J.-H.Y.)
| |
Collapse
|
10
|
Caban M, Fronik P, Terenzi A, Federa A, Bormio Nunes JH, Pitek R, Kirchhofer D, Schueffl HH, Berger W, Keppler BK, Kowol CR, Heffeter P. A new fluorescent oxaliplatin(iv) complex with EGFR-inhibiting properties for the treatment of drug-resistant cancer cells. Inorg Chem Front 2025; 12:1538-1552. [PMID: 39801772 PMCID: PMC11715172 DOI: 10.1039/d4qi03025g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025]
Abstract
Platinum chemotherapy is part of every second anticancer treatment regimen. However, its application is limited by severe side effects and drug resistance. The combination of platinum-based chemotherapeutics with EGFR inhibitors has shown remarkable synergism in clinical treatment. To enhance the tolerability of this combination, we designed a novel multi-action oxaliplatin-based platinum(iv) complex with an EGFR-inhibiting moiety (KP2749). KP2749 releases two independent cytotoxic agents upon reduction: oxaliplatin and the EGFR inhibitor KP2187, which was selected for its strong intrinsic fluorescence that became quenched upon complexation to metal ions. In particular, KP2749 demonstrated high stability and specific KP2187 release, with quenched fluorescent properties in its intact form, facilitating the investigation of its intracellular reduction. Notably, by exploiting its fluorescence, we demonstrated that intact KP2749 itself exhibited EGFR-inhibitory properties. Furthermore, subsequent experiments indicated that our complex was able to overcome resistance to oxaliplatin and EGFR inhibitors in vitro and in xenograft models in vivo. These effects were not only based on EGFR inhibition and DNA damage, but also improved cellular drug uptake. Finally, in silico docking analysis confirmed that the intact KP2749 complex had EGFR-binding properties, which were different from free KP2187. Consequently, these data suggested that the coordination of EGFR inhibitors to metal cores (like platinum) allow the fine-tuning of their EGFR-targeting properties. In conclusion, this study not only presents a new potential anticancer drug but also offers a novel fluorescent tool to study the intracellular drug release kinetics of platinum(iv) complexes.
Collapse
Affiliation(s)
- Monika Caban
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria +43 (0)1 40160-57557
| | - Philipp Fronik
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna Waehringer strasse 42 1090 Vienna Austria +43-4277-52680 +43-1-4277-52609
| | - Alessio Terenzi
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo Viale delle Scienze Ed. 17 90128 Palermo Italy
| | - Anja Federa
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna Waehringer strasse 42 1090 Vienna Austria +43-4277-52680 +43-1-4277-52609
- Vienna Doctoral School in Chemistry, University of Vienna Waehringer Strasse 42 1090 Vienna Austria
| | - Julia H Bormio Nunes
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria +43 (0)1 40160-57557
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna Waehringer strasse 42 1090 Vienna Austria +43-4277-52680 +43-1-4277-52609
| | - Rastislav Pitek
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria +43 (0)1 40160-57557
| | - Dominik Kirchhofer
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria +43 (0)1 40160-57557
| | - Hemma H Schueffl
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria +43 (0)1 40160-57557
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria +43 (0)1 40160-57557
- Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of, Vienna Austria
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna Waehringer strasse 42 1090 Vienna Austria +43-4277-52680 +43-1-4277-52609
- Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of, Vienna Austria
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna Waehringer strasse 42 1090 Vienna Austria +43-4277-52680 +43-1-4277-52609
- Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of, Vienna Austria
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria +43 (0)1 40160-57557
- Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of, Vienna Austria
| |
Collapse
|
11
|
Li X, Zhao X, Wang X, Xiong A, Wang Z, Shi Z, Zhang J, Wang H, Wei W, He C, Ma J, Guo Z, Duan C, Zhao J, Wang X. Programmable Modular Assembly of Homochiral Ir(III)-Metallohelices to Reverse Metallodrug Resistance by Inhibiting CDK1. Angew Chem Int Ed Engl 2025; 64:e202419292. [PMID: 39673540 DOI: 10.1002/anie.202419292] [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: 10/07/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/16/2024]
Abstract
Drug resistance is a major cause of cancer recurrence and poor prognosis. The innovative design and synthesis of inhibitors to target drug-resistance-specific proteins is highly desirable. However, challenges remain in precisely adjusting their conformation and stereochemistry to adapt the chiral regions of target proteins. Herein, using a stepwise programmable modular assembly approach, we precisely engineered two pairs of homochiral dinuclear Ir(III) metallohelices (Λ2S4-Hbpy and Δ2R4-Hbpy, Δ2S4-Hbpy and Λ2R4-Hbpy) functionalized with flexible dithiourea linkages. The resulting homochiral metallohelices exhibited significant chirality-dependent photocytotoxicities, and the enhanced structural compatibility of Δ2S4-Hbpy with the target cyclin-dependent kinase 1 (CDK1) contributed to its superior photodynamic therapy efficacy, achieving an outstanding photocytotoxicity index (PI) value of 2.3×104. Interestingly, emerging as a critical mediator in the development of oxaliplatin resistance, CDK1 targeting by Δ2S4-Hbpy achieved enhanced cellular uptake, anticancer activity, and oncosis-mediated cell death in oxaliplatin-resistant HCT-8/L cells. Mechanistic investigations, including proteomic profiling and CDK1 gene silencing, confirmed the pivotal role of chirality-selective CDK1 targeting in reversing metallodrug resistance. This study introduces a promising platform for constructing and customizing flexible metallohelices with precise conformation and stereochemistry to target drug-resistance-specific proteins, offering innovative insights into the designability of metallodrugs to overcome drug resistance.
Collapse
Affiliation(s)
- Xuezhao Li
- Cancer Hospital of Dalian University of Technology, School of Chemistry, State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Xing Zhao
- Cancer Hospital of Dalian University of Technology, School of Chemistry, State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Xingyun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Anxian Xiong
- Cancer Hospital of Dalian University of Technology, School of Chemistry, State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Zhicheng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Zhuolin Shi
- Cancer Hospital of Dalian University of Technology, School of Chemistry, State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Jingyi Zhang
- Chemistry and Biomedicine Innovation Center (ChemBIC), State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, China Institution
| | - Hanlin Wang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Wei Wei
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, China
| | - Cheng He
- Cancer Hospital of Dalian University of Technology, School of Chemistry, State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Jiajia Ma
- Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Chemistry and Chemical Engineering and Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Zijian Guo
- Chemistry and Biomedicine Innovation Center (ChemBIC), State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, China Institution
| | - Chunying Duan
- Cancer Hospital of Dalian University of Technology, School of Chemistry, State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Jing Zhao
- Chemistry and Biomedicine Innovation Center (ChemBIC), State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, China Institution
| | - Xiuxiu Wang
- Chemistry and Biomedicine Innovation Center (ChemBIC), State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, China Institution
| |
Collapse
|
12
|
Novohradsky V, Babu T, Kostrhunova H, Plaskow M, Markova L, Acharya S, Gibson D, Brabec V. Cisplatin-eugenol Pt(IV) prodrugs target colon cancer stem cells: A novel strategy for enhanced anticancer efficacy. Biomed Pharmacother 2025; 183:117854. [PMID: 39827811 DOI: 10.1016/j.biopha.2025.117854] [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: 10/11/2024] [Revised: 01/02/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Platinum(IV) compounds possess distinct properties that set them apart from platinum(II) compounds. Often designed as prodrugs, they are reduced within cancer cells to their active platinum(II) form, enabling their cytotoxic effects. Their versatility also lies in their ability to be functionalized and conjugated with bioactive molecules to enhance cancer cell targeting. This report introduces new prodrugs that combine antitumor cisplatin with axially coordinated eugenol, leveraging their synergistic action to target cancer stem cells. A third bioactive ligand, 4-phenylbutyrate or octanoate, was added to further enhance biological activity, creating 'triple action' prodrugs. These new platinum(IV) prodrugs offer a novel approach to cancer therapy by improving targeting, increasing efficacy, overcoming drug resistance, and reducing tumor invasiveness while sparing healthy tissue.
Collapse
Affiliation(s)
- Vojtech Novohradsky
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - Tomer Babu
- Institute for Drug Research, School of Pharmacy, The Hebrew University, Jerusalem 91120, Israel
| | - Hana Kostrhunova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - Menucha Plaskow
- Institute for Drug Research, School of Pharmacy, The Hebrew University, Jerusalem 91120, Israel
| | - Lenka Markova
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic
| | - Sourav Acharya
- Institute for Drug Research, School of Pharmacy, The Hebrew University, Jerusalem 91120, Israel
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University, Jerusalem 91120, Israel.
| | - Viktor Brabec
- Czech Academy of Sciences, Institute of Biophysics, Kralovopolska 135, Brno CZ-61200, Czech Republic; Department of Biophysics, Faculty of Science, Palacky University, Slechtitelu 27, Olomouc 783 71, Czech Republic.
| |
Collapse
|
13
|
Xuan Y, Yan Y, Wei X, Wang S, Zhang J, Tang Y, Li S. Positively-charged, chalcone-hydroxypyrone hybrid ruthenium(II)-arene complexes functionalized with ethacrynic acid: Synthesis, characterizaion, and antitumor effect. J Inorg Biochem 2025; 263:112778. [PMID: 39615317 DOI: 10.1016/j.jinorgbio.2024.112778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024]
Abstract
A new family of ethacrynic acid-functionalized, chalcone-hydroxypyrone hybrid ruthenium(II)-arene complexes (4a-4e) have been designed, synthesis and fully characterized by 1H and 13C NMR, ESI-MS, elemental analysis, and melting point tests. The molecular structure of 3a, one of the precursor complexes, has been determined by single-crystal X-ray diffraction. The cytotoxicity of the obtained complexes toward human cancer cell lines such as HeLa, MGC803, A549, MDA-MB-231, and MCF-7 cells have been investigated by MTT assay. Whereas complexes 4d and 4e showed significantly higher cytotoxicity than cisplatin (the positive control group) and complexes 3a-3e. Moreover, complexes 4d and 4e exhibited a certain selectivity (selectivity index: 7.33 and 7.57) toward MCF-7 cells over MCF-10a normal cells. Glutathione S-transferases (GSTs) activity assay indicate that complexes 4d and 4e exhibited higher GST inhibitory activity than ethacrynic acid (EA, the best characterized GST inhibitor), consistent with their higher cytotoxicity. Further mechanistic studies showed that 4e-induced cell apoptosis may be aroused by the production of ROS, the loss of mitochondrial membrane potential and G2/M phase cell arrest in MCF-7 cells. In addition, the in vivo antitumor effect study on the xenograft mouse models of MCF-7 cells reveal that complex 4e significantly inhibited tumor growth with a higher inhibition efficiency of 68.80 %, in comparison with the groups treated with cisplatin (59.25 %). These results highlight the strong possibility to develop positively-charged, chalcone-hydroxypyrone hybrid ruthenium(II)-arene complexes funcionalized with GST inhibitor as promising anticancer agents.
Collapse
Affiliation(s)
- Yuxin Xuan
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China
| | - Yuxi Yan
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China
| | - Xiaonan Wei
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China
| | - Shuxiang Wang
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China
| | - Jinchao Zhang
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China
| | - Yonghe Tang
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China.
| | - Shenghui Li
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China.
| |
Collapse
|
14
|
Fu H, Wang S, Gong Y, Dong H, Lai K, Yang Z, Fan C, Liu Z, Guo L. Triphenylphosphine-modified cyclometalated iridium III complexes as mitochondria-targeting anticancer agents with enhanced selectivity. Bioorg Chem 2025; 155:108148. [PMID: 39799728 DOI: 10.1016/j.bioorg.2025.108148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/29/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
This study presents the development and evaluation of triphenylphosphine-modified cyclometalated iridiumIII complexes as selective anticancer agents targeting mitochondria. By leveraging the mitochondrial localization capability of the triphenylphosphine group, these complexes displayed promising cytotoxicity in the micromolar range (3.12-7.24 μM) against A549 and HeLa cancer cells, these complexes exhibit significantly higher activity compared to their unmodified counterparts lacking the triphenylphosphine moiety. Moreover, they demonstrate improved specificity for cancer cells over normal cells, achieving selectivity index in the range of 5.46-14.83. Mechanistic studies confirmed that these complexes selectively target mitochondria rather than DNA, as shown by confocal microscopy and flow cytometry, where they accumulate to induce mitochondrial dysfunction. This disruption leads to mitochondrial membrane depolarization (MMP), elevated reactive oxygen species (ROS) levels, and activation of intrinsic apoptosis pathways. Furthermore, the complexes induce cell cycle arrest at the G2/M phase and suppress the migration of A549 cells.
Collapse
Affiliation(s)
- Hanxiu Fu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165 PR China
| | - Shuli Wang
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165 PR China
| | - Yuwen Gong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165 PR China
| | - Heqian Dong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165 PR China
| | - Kangning Lai
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165 PR China
| | - Zhihao Yang
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165 PR China
| | - Chunyan Fan
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165 PR China
| | - Zhe Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165 PR China.
| | - Lihua Guo
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165 PR China.
| |
Collapse
|
15
|
Niemeier F, Servos LM, Papadopoulos Z, Montesdeoca N, Ni K, Heinrich S, Karges J. Combinatorial Synthesis toward the Discovery of Highly Cytotoxic Fe(III) Complexes. J Med Chem 2025; 68:1316-1327. [PMID: 39680634 DOI: 10.1021/acs.jmedchem.4c01875] [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: 12/18/2024]
Abstract
Cancer remains one of the deadliest diseases worldwide, with some tumors proving difficult to treat and increasingly resistant to current therapies. Capitalizing on this, there is a need for new therapeutic agents with novel mechanisms of action. Among promising candidates, Fe(III) complexes have gained significant attention as potential chemotherapeutic agents. However, research on these compounds has been limited to a small number, leading to inefficiencies in drug discovery. This study addresses these limitations by developing a combinatorial library of 495 new Fe(III) complexes synthesized from aminophenol, hydroxybenzaldehyde, and pyridine derivatives. The compounds were screened for cytotoxicity against human breast adenocarcinoma and noncancerous fibroblasts, identifying a novel class of Fe(III) complexes with modest cancer cell selectivity. The lead compound effectively eradicated breast cancer tumor spheroids at low micromolar concentrations, highlighting the potential of this approach for rapid drug discovery.
Collapse
Affiliation(s)
- Felix Niemeier
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Lisa-Marie Servos
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Zisis Papadopoulos
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Nicolás Montesdeoca
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Kaixin Ni
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Sascha Heinrich
- Applied Microbiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| |
Collapse
|
16
|
Steel TR, Stjärnhage J, Lin Z, Bloomfield HO, Herbert CD, Astin JW, Krawczyk K, Rychlik B, Plażuk D, Jamieson SMF, Hartinger CG. Biotin functionalization of 8-hydroxyquinoline anticancer organometallics: low in vivo toxicity but potent in vitro activity. Dalton Trans 2025; 54:1583-1596. [PMID: 39659246 DOI: 10.1039/d4dt02296c] [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: 12/12/2024]
Abstract
[M(arene)(HQ)Cl] complexes (M = RuII/OsII/RhIII/IrIII; HQ = 8-hydroxyquinoline) have shown promise as anticancer agents. To assess the effect of conjugating biotin (vitamin B7) to such compounds and improve their tumor-targeting ability through interaction with the sodium-dependent multivitamin transporter (SMVT), the chlorido co-ligand was exchanged with biotinylated 6-aminoindazole. The complexes were characterized by NMR spectroscopy and mass spectrometry, and purity was determined by elemental analysis. The compounds were shown to be stable in aqueous solution but reacted in particular with biologically relevant nitrogen-donor ligands. The biotinylated organometallics were shown to be able to interact with the high-affinity biotin-binding protein streptavidin using molecular modelling. High antiproliferative activity of the biotinylated Rh complex (IC50 = 1.1-10 μM) and its chlorido precursor (IC50 = 2.1-7.0 μM) was demonstrated in human HCT116, NCI-H460, COLO 205, SW620, A2780 and A2780cis cancer cells, which feature differing levels of SMVT expression. While there was no clear relationship between the anticancer activity in cells and SMVT expression, the complexes showed similar activity in cisplatin-sensitive and -resistant cells. The most potent was the biotinylated Rh derivative which displayed low toxicity toward zebrafish embryos with >75% survival up to day 4 and after treatment with up to 32 μM complex.
Collapse
Affiliation(s)
- Tasha R Steel
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Julia Stjärnhage
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Zexiong Lin
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Hugh O Bloomfield
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Caitlin D Herbert
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jonathan W Astin
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Krzysztof Krawczyk
- Centre for Digital Biology and Biomedical Science - Biobank® Lodz, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| | - Błażej Rychlik
- Centre for Digital Biology and Biomedical Science - Biobank® Lodz, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| | - Damian Plażuk
- Laboratory of Molecular Spectroscopy, Department of Organic Chemistry, Faculty of Chemistry, University of Lodz, Tamka 12, 91-403 Łódź, Poland
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Christian G Hartinger
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
17
|
Mora-Lagos B, Reyes ME, Lobos-Gonzalez L, Del Campo M, Buchegger K, Zanella L, Riquelme I, Ili CG, Brebi P. Maraviroc/cisplatin combination inhibits gastric cancer tumoroid growth and improves mice survival. Biol Res 2025; 58:4. [PMID: 39827154 PMCID: PMC11748569 DOI: 10.1186/s40659-024-00581-3] [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: 05/09/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a significant cancer-related cause of death worldwide. GC's most used chemotherapeutic regimen is based on platinum drugs such as cisplatin (CDDP). However, CDDP chemoresistance reduces the survival rate of advanced GC. The immune C-C chemokine receptor type 5 (CCR5) have been proposed as a pivotal factor in cancer progression since its blockade has been linked with antineoplastic effects on tumor cell proliferation; nevertheless, its role in the chemoresistance of GC has not been elucidated. This study aimed to determine the effects induced by the CCR5 using Maraviroc (MVC), a highly selective CCR5 antagonist, on CDDP-resistant AGS cells (AGS R-CDDP), tumoroids (3D tumor spheroids), and animal models. RESULTS The combined CDDP and MVC treatment reduced cell viability and inhibited tumoroid formation in AGS R-CDDP cells. The effects of the MVC/CDDP combination on apoptosis and cell cycle progression were correlated with the increase in CDDP (dose-dependent). The mRNA levels of C-C Motif Chemokine Ligand 5 (CCL5), the main ligand for CCR5, decreased significantly in cells treated with the MVC/CDDP combination. MVC in the MVC/CDDP combination improved the survival rate and biochemical parameters of CDDP-treated mice by reducing the side effects of CDDP alone. CONCLUSIONS This finding suggests that MVC/CDDP combination could be a potential complementary therapy for GC.
Collapse
Affiliation(s)
- Bárbara Mora-Lagos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Temuco, Chile
| | - María Elena Reyes
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Temuco, Chile
| | - Lorena Lobos-Gonzalez
- Centro de Medicina Regenerativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
- Advanced Center for Chronic Diseases, ACCDiS, Santiago, Chile
- Laboratorio de comunicaciones celulares, Instituto de Ciencias Biomédicas, iCBM, Universidad de Chile, Santiago, Chile
| | - Matías Del Campo
- Centro de Medicina Regenerativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
- Advanced Center for Chronic Diseases, ACCDiS, Santiago, Chile
| | - Kurt Buchegger
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Biomedical Research Consortium-Chile (BMRC), Santiago, Chile
| | - Louise Zanella
- Doctorado en Ciencias Médicas, Universidad de La Frontera, Temuco, Chile
- Núcleo Milenio de Sociomedicina, Santiago, Chile
| | - Ismael Riquelme
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Temuco, Chile
| | - Carmen Gloria Ili
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Biomedical Research Consortium-Chile (BMRC), Santiago, Chile.
| | - Priscilla Brebi
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Biomedical Research Consortium-Chile (BMRC), Santiago, Chile.
| |
Collapse
|
18
|
Jin S, Feng C, Wang X. DNA or not DNA -that is the question determining the design of platinum anticancer drugs. Eur J Med Chem 2025; 282:117077. [PMID: 39579471 DOI: 10.1016/j.ejmech.2024.117077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/20/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024]
Abstract
Platinum drugs are the most widely used chemotherapeutics to treat various tumors. Their primary mode of action is supposed to be inducing apoptosis of cancer cells via covalent binding to DNA. This mechanism has shackled the design of new platinum drugs for many years. Mounting evidence shows that many platinum complexes form non-covalent adducts with DNA or interact with proteins to exhibit significant antitumor activity, thus implying some distinct mechanisms from that of traditional platinum drugs. These unconventional examples indicate that covalent DNA binding is not the precondition for the antitumor activity of platinum complexes, and diversified reactions or interactions with biomolecules, organelles, signal pathways, or immune system could lead to the antitumor activity of platinum complexes. The atypical mechanisms break the classical DNA-only paradigm and structure-activity relationships, thus opening a wide avenue for the design of innovative platinum anticancer drugs.
Collapse
Affiliation(s)
- Suxing Jin
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, PR China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Chenyao Feng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China.
| |
Collapse
|
19
|
Troisi R, Galardo F, Ferraro G, Lucignano R, Picone D, Marano A, Trifuoggi M, Sica F, Merlino A. Cisplatin/Apo-Transferrin Adduct: X-ray Structure and Binding to the Transferrin Receptor 1. Inorg Chem 2025; 64:761-765. [PMID: 39711171 DOI: 10.1021/acs.inorgchem.4c04435] [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: 12/24/2024]
Abstract
Here, we report the X-ray structure of the adduct formed upon reaction of cisplatin, one of the most prescribed anticancer agents for the clinic treatment of solid tumors, with the apo-form of human serum transferrin (hTF). Two Pt binding sites were identified in both molecules of the adduct present in the crystal asymmetric unit: Pt binds close to the side chains of Met256 and Met499 at the N- and C-lobe, respectively. In the crystal structure, the cisplatin moiety bound to Met256 also interacts with Ser616 from a symmetry related molecule. Structural analyses, together with in solution data, demonstrate that the presence of iron does not affect the ability of hTF to bind cisplatin and that the cisplatin binding does not significantly alter the overall conformation of the different forms of the protein that remain able to form a complex with the transferrin receptor 1 (TfR1). These data suggest that the different hTF forms can be used as nanocarriers for targeted (combined) metallodrug delivery.
Collapse
Affiliation(s)
- Romualdo Troisi
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Francesco Galardo
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Rosanna Lucignano
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Delia Picone
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Alessandra Marano
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Marco Trifuoggi
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Filomena Sica
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| |
Collapse
|
20
|
Li Y, Nie S, Wang L, Li D, Ma S, Li T, Sun H. Integrating deep learning in public health: a novel approach to PICC-RVT risk assessment. Front Public Health 2025; 12:1445425. [PMID: 39839389 PMCID: PMC11747573 DOI: 10.3389/fpubh.2024.1445425] [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: 06/07/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Background Machine learning is pivotal for predicting Peripherally Inserted Central Catheter-related venous thrombosis (PICC-RVT) risk, facilitating early diagnosis and proactive treatment. Existing models often assess PICC-RVT risk as static and discrete outcomes, which may limit their practical application. Objectives This study aims to evaluate the effectiveness of seven diverse machine learning algorithms, including three deep learning and four traditional machine learning models, that incorporate time-series data to assess PICC-RVT risk. It also seeks to identify key predictive factors for PICC-RVT using these models. Methods We conducted a retrospective multi-center cohort study involving 5,272 patients who underwent PICC placement. After preprocessing patient data, the models were trained. Demographic, clinical pathology, and treatment data were analyzed to identify predictive factors. A variable analysis was then conducted to determine the most significant predictors of PICC-RVT. Model performance was evaluated using the Concordance Index (c-index) and the composite Brier score, and the Intraclass Correlation Coefficient (ICC) from cross-validation folds assessed model stability. Results Deep learning models generally outperformed traditional machine learning models in terms of predictive accuracy (mean c-index: 0.949 vs. 0.732; mean integrated Brier score: 0.046 vs. 0.093). Specifically, the DeepSurv model demonstrated exceptional precision in risk assessment (c-index: 0.95). Stability varied with the number of predictive factors, with Cox-Time showing the highest ICC (0.974) with 16 predictive factors, and DeepSurv the most stable with 26 predictive factors (ICC: 0.983). Key predictors across models included albumin levels, prefill sealant type, and activated partial thromboplastin time. Conclusion Machine learning models that incorporate time-to-event data can effectively predict PICC-RVT risk. The DeepSurv model, in particular, shows excellent discriminative and calibration capabilities. Albumin levels, type of prefill sealant, and activated partial thromboplastin time are critical indicators for identifying and managing high-risk PICC-RVT patients.
Collapse
Affiliation(s)
- Yue Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Electronic and Information Engineering, TianGong University, Tianjin, China
| | - Shengxiao Nie
- Department of Nursing, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lei Wang
- Department of Nursing, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Dongsheng Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shengmiao Ma
- School of Nursing, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Sun
- Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Gai S, Meng L, Qin Y, Jiang M. Synthesis and Anticancer Studies of Pt(II) Complex Derived From 4-Phenylthiosemicarbazone. Chem Biodivers 2025:e202402972. [PMID: 39745361 DOI: 10.1002/cbdv.202402972] [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: 11/11/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025]
Abstract
Although cisplatin is widely used as a first-line chemotherapy agent, it has significant side effects. Herein, we synthesized a Pt(II) complex (Pt1) derived from o-vanillin-4-phenylthiosemicarbazone ligand and confirmed its crystal structure by x-ray crystallography. Complex Pt1 exhibited potent anticancer activity against various tested cancer cell lines, with particular efficacy against HepG-2 cells. Further investigations revealed that Pt1 inhibited the growth of HepG-2 cells through multiple mechanisms, including the generation of excessive reactive oxygen species (ROS), induction of DNA damage, enhancement of mitochondrial membrane permeability, promotion of apoptosis, and activation of autophagic cell death.
Collapse
Affiliation(s)
- Shuangshuang Gai
- Institute for History and Culture of Science & Technology, Guangxi Minzu University, Nanning, Guangxi, China
| | - Lili Meng
- School of Biological and Food engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi, China
| | - Yiming Qin
- School of Biological and Food engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi, China
| | - Ming Jiang
- School of Biological and Food engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi, China
| |
Collapse
|
22
|
Lin JF, Liu ZX, Chen DL, Huang RZ, Cao F, Yu K, Li T, Mo HY, Sheng H, Liang ZB, Liao K, Han Y, Li SS, Zeng ZL, Gao S, Ju HQ, Xu RH. Nucleus-translocated GCLM promotes chemoresistance in colorectal cancer through a moonlighting function. Nat Commun 2025; 16:263. [PMID: 39747101 PMCID: PMC11696352 DOI: 10.1038/s41467-024-55568-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Metabolic enzymes perform moonlighting functions during tumor progression, including the modulation of chemoresistance. However, the underlying mechanisms of these functions remain elusive. Here, utilizing a metabolic clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 knockout library screen, we observe that the loss of glutamate-cysteine ligase modifier subunit (GCLM), a rate-limiting enzyme in glutathione biosynthesis, noticeably increases the sensitivity of colorectal cancer (CRC) cells to platinum-based chemotherapy. Mechanistically, we unveil a noncanonical mechanism through which nuclear GCLM competitively interacts with NF-kappa-B (NF-κB)-repressing factor (NKRF), to promote NF-κB activity and facilitate chemoresistance. In response to platinum drug treatment, GCLM is phosphorylated by P38 MAPK at T17, resulting in its recognition by importin a5 and subsequent nuclear translocation. Furthermore, elevated expression of nuclear GCLM and phospho-GCLM correlate with an unfavorable prognosis and poor benefit from standard chemotherapy. Overall, our work highlights the essential nonmetabolic role and posttranslational regulatory mechanism of GCLM in enhancing NF-κB activity and subsequent chemoresistance.
Collapse
Affiliation(s)
- Jin-Fei Lin
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Ze-Xian Liu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Dong-Liang Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Ren-Ze Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Fen Cao
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, PR China
| | - Kai Yu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ting Li
- Department of Gastroenterology and Urology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
| | - Hai-Yu Mo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Hui Sheng
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Zhi-Bing Liang
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Kun Liao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yi Han
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Shan-Shan Li
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Zhao-Lei Zeng
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Song Gao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Huai-Qiang Ju
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China.
| | - Rui-Hua Xu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, PR China.
| |
Collapse
|
23
|
Wang Z, Kregel M, Meijers JL, Franch J, Cuijpers VMJI, Ahlers D, Karst U, Slootweg P, van der Geest IC, Leeuwenburgh SC, van den Beucken JJ. Cisplatin-functionalized dual-functional bone substitute granules for bone defect treatment after bone tumor resection. Acta Biomater 2025; 191:158-176. [PMID: 39551330 DOI: 10.1016/j.actbio.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Invasive bone tumors pose a significant healthcare challenge, often requiring systemic chemotherapy and limb salvage surgery. However, these strategies are hampered by severe side effects, complex post-resection bone defects, and high local recurrence rates. To address this, we developed dual-functional bone substitute biomaterials by functionalizing commercially available bone substitute granules (Bio-Oss® and MBCP®+) with the established anticancer agent cisplatin. Physicochemical characterization revealed that Bio-Oss® granules possess a higher surface area and lower crystallinity compared to MBCP®+ granules, which enhances their capacity for cisplatin adsorption and release. In co-cultures with metastatic breast and prostate cancer cells (MDA-MB-231 and PC3) and bone marrow stromal cells (hBMSCs), cisplatin-functionalized granules and their releasates exhibited dose-dependent cytotoxic effects on cancer cells while having less impact on hBMSCs. Furthermore, investigations on the mechanism of action indicated that cisplatin induced significant cell cycle arrest and apoptosis in MDA-MB-231 and PC3 cells, contrasting with minimal effects on hBMSCs. In a rat femoral condyle defect model, cisplatin-functionalized granules did not evoke adverse effects on bone tissue ingrowth or new bone formation. Importantly, local application of cisplatin-functionalized granules resulted in negligible cisplatin accumulation without signs of apoptotic damage in kidneys and livers. Taken together, we here provide hard evidence that cisplatin-functionalized granules maintain a favorable balance between biosafety, anticancer efficacy, and bone regenerative capacity. Consequently, loading granular bone substitutes with cisplatin holds promise for local treatment of bone defects following bone tumor resections, presenting a safe and potentially more effective alternative to systemic cisplatin administration. STATEMENT OF SIGNIFICANCE: Current treatments in combating malignant bone tumors are hampered by severe side effects, high local tumor recurrence, and complex bone defects after surgery. This study explores a facile manufacturing method to render two types of commercially available bone substitute granules (Bio-Oss® and MBCP®+) suitable for local delivery of cisplatin. The use of cisplatin-functionalized granules has shown promising results both in killing cancer cells in a dose-dependent manner and in aiding bone regeneration. Importantly, this local treatment strategy avoids the systemic toxicity associated with traditional chemotherapy to excretory organs. This dual-functional strategy represents a significant advancement in bone cancer treatment, offering a safe and more efficient alternative that could improve outcomes for patients following bone tumor resection.
Collapse
Affiliation(s)
- Zhule Wang
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands
| | - Mark Kregel
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - Jean-Luc Meijers
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - Jordi Franch
- Department of Small Animal Medicine and Surgery, Veterinary School, Universitat Autonoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Vincent M J I Cuijpers
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - David Ahlers
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Piet Slootweg
- Department of Pathology, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, the Netherlands
| | - Ingrid Cm van der Geest
- Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands; Department of Orthopedics, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, the Netherlands
| | - Sander Cg Leeuwenburgh
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands
| | - Jeroen Jjp van den Beucken
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands.
| |
Collapse
|
24
|
Dómine Gómez M, Subbiah V, Peters S, Sala MA, Trigo J, Paz-Ares L, Nieto Archilla A, Gomez Garcia J, Alvarez García C, López-Vilariño de Ramos JA, Kahatt Lopez C, Fernandez CM. Lurbinectedin is an effective alternative to platinum rechallenge and may restore platinum sensitivity in patients with sensitive relapsed small cell lung cancer. Expert Rev Anticancer Ther 2025; 25:27-40. [PMID: 39660812 DOI: 10.1080/14737140.2024.2438067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
INTRODUCTION Platinum rechallenge is recommended for patients with small cell lung cancer (SCLC) who relapse ≥90 days after completing first-line chemotherapy, although it may not always be the most suitable option. AREAS COVERED Articles for review were identified via PubMed and ClinicalTrials.gov searches, supplemented with non-indexed publications (e.g. conference abstracts) known to the manufacturer. We examined evidence for platinum re-exposure in patients with sensitive relapsed SCLC, and present lurbinectedin as a potential alternative. The complementary mechanisms of action of lurbinectedin and platinum, owing to opposite sensitivity of SCLC cells, may resensitize tumor cells to platinum. As efficacy outcomes with lurbinectedin are equivalent or better than those with platinum rechallenge and its hematological safety profile is more favorable, achieving maximum dose intensity is more likely. The simpler dosing schedule of lurbinectedin (1 vs 3 days) and lack of need for granulocyte colony-stimulating factor primary prophylaxis lessens treatment burden. EXPERT OPINION Incorporation of lurbinectedin into therapeutic algorithms for relapsed SCLC has challenged long-established treatment paradigms. Initial evidence indicates that using lurbinectedin after failure of first-line platinum may prolong the platinum-free interval and reserve platinum for later use. Current evidence supports lurbinectedin as a second-line option in patients with sensitive relapsed SCLC.
Collapse
Affiliation(s)
- Manuel Dómine Gómez
- Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Vivek Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, TN, USA
| | - Solange Peters
- Oncology Department - CHUV, Lausanne University, Lausanne, Switzerland
| | - María Angeles Sala
- Medical Oncology Department, Hospital Universitario Basurto, Bilbao, Spain
| | - José Trigo
- Medical Oncology Department, Hospital HC Marbella, Málaga, Spain
| | - Luis Paz-Ares
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
25
|
Hsu CY, Lin J, Wei MF, Chen LH, Liang HKT, Lin FH. Local delivery of carboplatin-loaded hydrogel and calcium carbonate enables two-stage drug release for limited-dose radiation to eliminate mouse malignant glioma. Biomaterials 2025; 312:122746. [PMID: 39106816 DOI: 10.1016/j.biomaterials.2024.122746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/07/2023] [Accepted: 08/02/2024] [Indexed: 08/09/2024]
Abstract
Postoperative radiotherapy remains the gold standard for malignant glioma treatment. Clinical limitations, including tumor growth between surgery and radiotherapy and the emergence of radioresistance, reduce treatment effectiveness and result in local disease progression. This study aimed to develop a local drug delivery system to inhibit tumor growth before radiotherapy and enhance the subsequent anticancer effects of limited-dose radiotherapy. We developed a compound of carboplatin-loaded hydrogel (CPH) incorporated with carboplatin-loaded calcium carbonate (CPCC) to enable two-stage (peritumoral and intracellular) release of carboplatin to initially inhibit tumor growth and to synergize with limited-dose radiation (10 Gy in a single fraction) to eliminate malignant glioma (ALTS1C1 cells) in a C57BL/6 mouse subcutaneous tumor model. The doses of carboplatin in CPH and CPCC treatments were 150 μL (carboplatin concentration of 5 mg/mL) and 15 mg (carboplatin concentration of 4.1 μg/mg), respectively. Mice receiving the combination of CPH-CPCC treatment and limited-dose radiation exhibited significantly reduced tumor growth volume compared to those receiving double-dose radiation alone. Furthermore, combining CPH-CPCC treatment with limited-dose radiation resulted in significantly longer progression-free survival than combining CPH treatment with limited-dose radiation. Local CPH-CPCC delivery synergized effectively with limited-dose radiation to eliminate mouse glioma, offering a promising solution for overcoming clinical limitations.
Collapse
Affiliation(s)
- Cheng-Yi Hsu
- Department of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei 10617, Taiwan.
| | - Jason Lin
- Department of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei 10617, Taiwan.
| | - Ming-Feng Wei
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, No. 7, Chung Shan South Rd., Zhongzheng Dist., Taipei 10002, Taiwan.
| | - Liang-Hsin Chen
- Department of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei 10617, Taiwan; Division of Proton Therapy, Department of Radiation Oncology, National Taiwan University Cancer Center, National Taiwan University College of Medicine, No.57, Ln. 155, Sec. 3, Keelung Rd., Da'an Dist., Taipei 10672, Taiwan.
| | - Hsiang-Kuang Tony Liang
- Department of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei 10617, Taiwan; Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, No. 7, Chung Shan South Rd., Zhongzheng Dist., Taipei 10002, Taiwan; Division of Proton Therapy, Department of Radiation Oncology, National Taiwan University Cancer Center, National Taiwan University College of Medicine, No.57, Ln. 155, Sec. 3, Keelung Rd., Da'an Dist., Taipei 10672, Taiwan.
| | - Feng-Huei Lin
- Department of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei 10617, Taiwan; Institute of Biomedical Engineering and Nano-medicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, Miaoli County 35053, Miaoli County, Taiwan.
| |
Collapse
|
26
|
Antonets AA, Spitsyna EV, Tyurin VY, Mazur DM, Yakovlev DS, Babkov DA, Pshenichnikova MS, Spasov AA, Milaeva ER, Nazarov AA. Ruthenium complexes with abiraterone acetate as antiproliferative agents. J Inorg Biochem 2025; 262:112754. [PMID: 39383670 DOI: 10.1016/j.jinorgbio.2024.112754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
This study is dedicated to the development of multimodal anticancer agents. We have obtained ruthenium complexes conjugated with the steroid-type antitumor drug abiraterone acetate in order to take advantage of the dual antitumor properties of both ruthenium and abiraterone. The compounds exhibit good antiproliferative activity against cancer cells, with selectivity over primary fibroblasts. Real-time cell analysis revealed that compound dichlorido(η6-p-cymene)(abiraterone acetate)ruthenium(II) had pronounced antiproliferation activity compared to abiraterone acetate. Flow cytometric studies on the mechanism of cell death have revealed that the most active compound induces apoptosis more effectively than abiraterone acetate. Our findings demonstrate the potential of this novel dual-action compound as promising candidates for further development as anticancer agents.
Collapse
Affiliation(s)
- Anastasia A Antonets
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Ekaterina V Spitsyna
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Vladimir Yu Tyurin
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia; Department of Materials Science, MSU-BIT University, Shenzhen 517182, China
| | - Dmitrii M Mazur
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Dmitry S Yakovlev
- Volgograd State Medical University, Pavshikh Bortsov Sq. 1, 400131 Volgograd, Russia
| | - Denis A Babkov
- Volgograd State Medical University, Pavshikh Bortsov Sq. 1, 400131 Volgograd, Russia
| | | | - Alexander A Spasov
- Volgograd State Medical University, Pavshikh Bortsov Sq. 1, 400131 Volgograd, Russia
| | - Elena R Milaeva
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Alexey A Nazarov
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia.
| |
Collapse
|
27
|
Taguchi H, Sumi D, Uemura A, Matsumoto K, Fujishiro H. Cisplatin caused highly delayed cytotoxicity in the immortalized cells derived from S3 segment of mouse kidney proximal tubules. Toxicol Appl Pharmacol 2025; 494:117171. [PMID: 39592085 DOI: 10.1016/j.taap.2024.117171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
Anti-cancer drug cisplatin (CDDP) causes severe acute kidney injury (AKI). CDDP-induced AKI does not occur immediately after administration, but rather 6 to 10 days after administration. However, the mechanism underling the delayed renal injury by CDDP is not well understood. In a previous investigation using immortalized cells derived from the S1, S2, and S3 segments of the proximal tubules, we found that S3 cells were more sensitive to CDDP than S1 and S2 cells. In this study, we examined whether S1, S2, and S3 cells would be useful in elucidating the mechanism of CDDP-induced delayed renal injury and whether the high sensitivity of S3 cells contributes to CDDP-induced delayed renal injury. Measurement of platinum (Pt) content by ICP-MS showed that Pt accumulation peaked at 15 min after CDDP exposure in each cell type. Even when the medium was replaced with CDDP-free medium after the 15-min CDDP exposure and the cells were further incubated, delayed cytotoxicity was still observed. The S3 cells exhibited greater sensitivity to CCDP than the S1 and S2 cells at all time points after the medium change. To investigate the mechanism of the CDDP-induced delayed cytotoxicity, we examined the cell cycle distribution of cells after CDDP exposure. The results showed that CDDP-induced perturbation of cell cycle was greater in S3 than in S1 and S2 cells. These results suggest that perturbation of the cell cycle in S3 cells due to enhanced CDDP-DNA adduct formation contributes to the high susceptibility of S3 cells to CDDP-induced delayed cytotoxicity.
Collapse
Affiliation(s)
- Hiroki Taguchi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Daigo Sumi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Ayumi Uemura
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Kanako Matsumoto
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Hitomi Fujishiro
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| |
Collapse
|
28
|
Delinassios JG, Hoffman RM, Koumakis G, Palitskaris D, Poulatsidou KN, Delinasios GJ. Sub-toxic cisplatin concentrations induce extensive chromosomal, nuclear and nucleolar abnormalities associated with high malignancy before acquired resistance develops: Implications for clinical caution. PLoS One 2024; 19:e0311976. [PMID: 39724069 DOI: 10.1371/journal.pone.0311976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/27/2024] [Indexed: 12/28/2024] Open
Abstract
AIM This study investigates the impact of sub-toxic cisplatin levels on nuclear and nucleolar abnormalities and chromosome instability in HeLa cells since our current knowledge of cisplatin effects on these parameters is based on studies with high concentrations of cisplatin. MATERIALS AND METHODS HeLa cells were exposed to gradually increasing sub-toxic doses of cisplatin (0.01 to 0.2 μg/ml). Cells treated with 0.1 and 0.2 μg/ml, termed HeLaC0.1 and HeLaC0.2, were not cisplatin-resistant, only exhibiting a slightly reduced viability, and were termed "cisplatin-sensitized cells." Giemsa and silver staining were used to detect nuclear and nucleolar abnormalities and chromosomal alterations. RESULTS Notable abnormalities were observed in HeLaC0.1 and HeLaC0.2 cells after treatment with sub-toxic concentrations of cisplatin: nuclei showed abnormal shapes, blebs, micronuclei, fragmentation, pulverization, and multinucleation; nucleoli exhibited irregular shapes and increased numbers; anaphase cells showed more nucleolar organizing regions. Abnormal chromosome segregation, heightened aneuploidy (81-140 chromosomes), polyploidy, double minutes, dicentrics, chromatid exchanges, chromatid separations, pulverization, and chromosome markers were prominently noted. These abnormalities were intensified in cells pre-sensitized to 0.02 or 0.08 μg/ml cisplatin for seven days, then exposed to 0.03 or 0.1 μg/ml cisplatin for 24 hours, and finally cultured in cisplatin-free medium for 24 hours before chromosome analysis. CONCLUSION HeLa cells subjected to increasing concentrations of sub-toxic cisplatin exhibited large-scale, multiple-type abnormalities in nuclei, nucleoli, chromosomes, and chromosomal numbers, indicating genetic/chromosomal instability associated with high malignancy, before the development of cisplatin resistance. These results suggest that low doses of cisplatin administration in the clinical setting may promote malignancy and caution should be used with this type of treatment.
Collapse
Affiliation(s)
- John G Delinassios
- International Institute of Anticancer Research, Kapandriti, Attica, Greece
| | - Robert M Hoffman
- Department of Surgery, University of California, La Jolla, California, United States of America
- AntiCancer Inc., San Diego, San Diego, California, United States of America
| | - George Koumakis
- International Institute of Anticancer Research, Kapandriti, Attica, Greece
| | | | | | | |
Collapse
|
29
|
Airik M, Clayton K, Wipf P, Airik R. JP4-039 Mitigates Cisplatin-Induced Acute Kidney Injury by Inhibiting Oxidative Stress and Blocking Apoptosis and Ferroptosis in Mice. Antioxidants (Basel) 2024; 13:1534. [PMID: 39765862 PMCID: PMC11727076 DOI: 10.3390/antiox13121534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/15/2025] Open
Abstract
Cisplatin is a commonly used chemotherapeutic agent in the treatment of a wide array of cancers. Due to its active transport into the kidney proximal tubule cells, cisplatin treatment can cause a buildup of this nephrotoxic compound in the kidney, resulting in acute kidney injury (AKI). About 30% of patients receiving cisplatin chemotherapy develop cisplatin-induced AKI. JP4-039 is a mitochondria-targeted reactive oxygen species (ROS) and electron scavenger. Recent studies have shown that JP4-039 mitigates a variety of genotoxic insults in preclinical studies in rodents by suppressing oxidative stress-mediated tissue damage and blocking apoptosis and ferroptosis. However, the benefits of JP4-039 treatment have not been tested in the setting of AKI. In this study, we investigated the potential renoprotective effect of JP4-039 on cisplatin-induced AKI. To address this goal, we treated mice with JP4-039 before or after cisplatin administration and analyzed them for functional and molecular changes in the kidney. JP4-039 co-administration attenuated cisplatin-induced renal dysfunction and histopathological changes. Upregulation of tubular injury markers was also suppressed by JP4-039. Mechanistically, JP4-039 suppressed lipid peroxidation, prevented tissue oxidative stress, and preserved the glutathione levels in cisplatin-injected mice. An increase in cisplatin-induced apoptosis and ferroptosis was also alleviated by the compound. Moreover, JP4-039 inhibited cytokine overproduction in cisplatin-injected mice. Together, our findings demonstrate that JP4-039 is a promising therapeutic agent against cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Merlin Airik
- Division of Nephrology, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Kacian Clayton
- Division of Nephrology, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Rannar Airik
- Division of Nephrology, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
30
|
Wang M, Li F, Wang Z, Lv L, Liu W. Research progress of natural product-conjugated platinum and gold complexes as potential antitumor agents. Eur J Med Chem 2024; 280:116956. [PMID: 39413444 DOI: 10.1016/j.ejmech.2024.116956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Cancer is widely recognized as a serious disease that poses a significant threat to human life and health. The distinctive chemical properties and pronounced antiproliferative activity of platinum drugs are considered to be responsible for their remarkable efficacy in clinical applications. However, undesirable side effects and resistance have severely hampered the treatment of various types of cancer with platinum-based drugs. Natural products (NPs) exhibit extensive pharmacological activities and represent an important source for developing cancer therapeutics. Therefore, the combination of metals and NPs is an attractive strategy for the development of new anticancer agents. Several studies have indicated that combining metals with NPs has a synergistic enhancement effect in antitumor activity. For transition metals, there has been burgeoning research output investigating NP-conjugated platinum and gold complexes. The present article reviews the progress made over the past 5-10 years on the development of NP-conjugated platinum and gold complexes, including a brief introduction to their chemistry and mechanism of action, and a summary of their benefits.
Collapse
Affiliation(s)
- Meiyu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Fuwei Li
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zhaoran Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Lin Lv
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Wukun Liu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
31
|
Audzeyenka I, Piwkowska A, Rogacka D, Makowski M, Kowalik M. Biological Evaluation of a Rhodium(III) Bipyridylsulfonamide Complex: Effects on Mitochondrial Dynamics and Cytoskeletal Remodeling in Breast Cancer Cells. J Med Chem 2024; 67:21364-21379. [PMID: 39576967 DOI: 10.1021/acs.jmedchem.4c02284] [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: 11/24/2024]
Abstract
Rhodium(III) complexes have gained attention for their anticancer potential. In this study, we investigated a rhodium(III) bipyridylsulfonamide complex (2) and its ligand (L) for their effects on breast cancer (SKBr3) and noncancerous mammary cells (HB2). Both compounds significantly reduced oxidative phosphorylation (OXPHOS) and mitochondrial function in SKBr3 cells while sparing HB2 cells. Compound 2 also increased glycolysis in both lines, suggesting a metabolic shift. Mitochondrial size and shape were altered, particularly in SKBr3 cells. Additionally, both compounds reduced cancer cell migration by disrupting actin cytoskeleton organization and the Rac1/VASP signaling pathway. These findings suggest that the rhodium(III) bipyridylsulfonamide complex selectively impairs mitochondrial dynamics and cell migration in cancer cells while sparing healthy cells, providing insight into its mechanism of action and toward its use as targeted anticancer therapy. This study lays the groundwork for future in vivo studies and further optimization of these metal-based therapeutics for clinical applications.
Collapse
Affiliation(s)
- Irena Audzeyenka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, W. Stwosza 63, 80-308 Gdansk, Poland
| | - Agnieszka Piwkowska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, W. Stwosza 63, 80-308 Gdansk, Poland
| | - Dorota Rogacka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, W. Stwosza 63, 80-308 Gdansk, Poland
| | - Mariusz Makowski
- Faculty of Chemistry, Department of Bioinorganic Chemistry, University of Gdańsk, W. Stwosza 63, 80-308 Gdańsk, Poland
| | - Mateusz Kowalik
- Faculty of Chemistry, Department of Bioinorganic Chemistry, University of Gdańsk, W. Stwosza 63, 80-308 Gdańsk, Poland
| |
Collapse
|
32
|
Biswas M, Chaudhary K, Padhi SS, Banerjee A, Bharathavikru RS, Bandaru S, Panda SJ, Purohit CS, Das NR, Pathak RK. TTFA-Platin Conjugate: Deciphering the Therapeutic Roles of Combo-Prodrug through Evaluating Stability-Activity Relationship. J Med Chem 2024; 67:20986-21008. [PMID: 39611754 DOI: 10.1021/acs.jmedchem.4c01545] [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: 11/30/2024]
Abstract
This work introduces a novel Pt(II) based prodrug TTFA-Platin that integrates a β-diketonate ligand TTFA with a platinum scaffold to structurally resemble carboplatin and offers intermediate kinetic lability between cisplatin and carboplatin, striking a balance between therapeutic efficacy and safety. A comprehensive stability and speciation study was conducted in various biological media, mapping the therapeutic effects of TTFA-Platin. A control molecule, TMK-Platin, was synthesized to further validate the structural-stability relationship, which displayed poor activatable features in biological systems. In vitro studies against a panel of cancer cell lines revealed that TTFA-Platin exhibited significantly higher potency compared to TMK-Platin. In vivo studies revealed that TTFA-Platin exhibited significantly lower toxicity than the reference platinum compounds. Thus, leveraging ligands that fine-tune kinetic lability and offer therapeutic benefits can help develop more effective and safer cancer treatments, addressing the limitations of existing therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Sateesh Bandaru
- Key Laboratory of Novel Materials for Sensor of Zhejiang Province College of Materials and Environmental Engineering, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Subhra Jyoti Panda
- School of Chemical Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar 752050, India
| | - Chandra Shekhar Purohit
- School of Chemical Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar 752050, India
| | - Nihar Ranjan Das
- Roland Institute of Pharmaceutical Sciences, Berhampur 760010, Odisha, India
| | | |
Collapse
|
33
|
Cao Z, Yan R, Chen J, She M, Jia S, Sun W, Liu P, Zhang S, Li JL. Water-Soluble Fluorescent Sensors for Quantification of Trace Cisplatin in Body Fluids from Clinical Cancer Patients. J Am Chem Soc 2024; 146:33651-33662. [PMID: 39607057 DOI: 10.1021/jacs.4c10460] [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: 11/29/2024]
Abstract
Accurate quantification of cisplatin (cDDP) in body fluids (blood, urine, and ascites) is crucial in monitoring therapeutic processes, assessing drug metabolism, and optimizing treatment schedules for cancer patients. Nonetheless, due to the inherent fluorescence and complexity of the body fluid matrix, along with the low cDDP concentrations in these fluids during treatment, using fluorescent sensors for fluid detection remains a subject of ongoing research. Herein, a series of water-soluble cDDP-activatable fluorescent sensors was rationally constructed by introducing thioether groups to the xanthene skeleton based on the chalcogenophilicity of platinum. These sensors exhibit excellent sensitivity and certain anti-interference capabilities for sensing cDDP in living cells, rat tissues, and zebrafish. Especially, with a simplified sample pretreatment procedure, for the first time, Rh3 and Rh4 have enabled quantitative detection of cDDP levels in diversiform body fluids from clinical ovarian and bladder cancer patients. These results are highly consistent with those obtained by ICP-MS detection. This work paves the way for utilizing fluorescent sensors in clinical body fluid analysis, thus potentially revolutionizing the monitoring methods of cDDP in clinic settings.
Collapse
Affiliation(s)
- Zifeng Cao
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, Xi'an Key Laboratory of Functional Supramolecular Structure and Materials, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi Province 710127, China
| | - Rong Yan
- The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi Province 710061, China
| | - Jiao Chen
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, Xi'an Key Laboratory of Functional Supramolecular Structure and Materials, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi Province 710127, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Lab of Tissue Engineering, the College of Life Sciences, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi Province 710069, China
| | - Mengyao She
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, Xi'an Key Laboratory of Functional Supramolecular Structure and Materials, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi Province 710127, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Lab of Tissue Engineering, the College of Life Sciences, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi Province 710069, China
- Shaanxi Key Laboratory for Carbon Neutral Technology, Carbon Neutrality College (YuLin), Northwest University, Yulin, Shaanxi Province 719099, China
| | - Shanshan Jia
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, Xi'an Key Laboratory of Functional Supramolecular Structure and Materials, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi Province 710127, China
| | - Wei Sun
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, Xi'an Key Laboratory of Functional Supramolecular Structure and Materials, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi Province 710127, China
| | - Ping Liu
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, Xi'an Key Laboratory of Functional Supramolecular Structure and Materials, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi Province 710127, China
| | - Shengyong Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, Xi'an Key Laboratory of Functional Supramolecular Structure and Materials, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi Province 710127, China
| | - Jian-Li Li
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, Xi'an Key Laboratory of Functional Supramolecular Structure and Materials, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi Province 710127, China
- Shaanxi Key Laboratory for Carbon Neutral Technology, Carbon Neutrality College (YuLin), Northwest University, Yulin, Shaanxi Province 719099, China
| |
Collapse
|
34
|
Lou J, Wu F, He W, Hu R, Cai Z, Chen G, Zhao W, Zhang Z, Si Y. Hesperidin activates Nrf2 to protect cochlear hair cells from cisplatin-induced damage. Redox Rep 2024; 29:2341470. [PMID: 38629504 PMCID: PMC11025410 DOI: 10.1080/13510002.2024.2341470] [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: 04/19/2024] Open
Abstract
Cisplatin is widely employed in clinical oncology as an anticancer chemotherapy drug in clinical practice and is known for its severe ototoxic side effects. Prior research indicates that the accumulation of reactive oxygen species (ROS) plays a pivotal role in cisplatin's inner ear toxicity. Hesperidin is a flavanone glycoside extracted from citrus fruits that has anti-inflammatory and antioxidant effects. Nonetheless, the specific pharmacological actions of hesperidin in alleviating cisplatin-induced ototoxicity remain elusive. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a critical mediator of the cellular oxidative stress response, is influenced by hesperidin. Activation of Nrf2 was shown to have a protective effect against cisplatin-induced ototoxicity. The potential of hesperidin to stimulate Nrf2 in attenuating cisplatin's adverse effects on the inner ear warrants further investigation. This study employs both in vivo and in vitro models of cisplatin ototoxicity to explore this possibility. Our results reveal that hesperidin mitigates cisplatin-induced ototoxicity by activating the Nrf2/NQO1 pathway in sensory hair cells, thereby reducing ROS accumulation, preventing hair cell apoptosis, and alleviating hearing loss.
Collapse
Affiliation(s)
- Jintao Lou
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Fan Wu
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Wuhui He
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Rui Hu
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Ziyi Cai
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Guisheng Chen
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Wenji Zhao
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Zhigang Zhang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yu Si
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
35
|
Zhu J, Yuan A, Le Y, Chen X, Guo J, Liu J, Chen H, Wang CY, Lu D, Lu K. Yi-Qi-Jian-Pi-Xiao-Yu formula inhibits cisplatin-induced acute kidney injury through suppressing ferroptosis via STING-NCOA4-mediated ferritinophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156189. [PMID: 39515100 DOI: 10.1016/j.phymed.2024.156189] [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: 06/04/2024] [Revised: 09/28/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The kidneys are the primary excretory organs for platinum drugs, making them susceptible to damage from these drugs. Cisplatin-induced acute kidney injury (CIAKI) is the most common side effect observed in patients undergoing clinical cisplatin treatment. A traditional Chinese medicinal preparation, the Yi-Qi-Jian-Pi-Xiao-Yu formula (YQJPXY), which is a modified formulation of the classical Chinese medicine formula Buyang Huanwu Decoction, has long been used in the treatment of clinical kidney diseases. It is expected to be used to ameliorate cisplatin-induced acute kidney injury. However, the mechanism of this YQJPXY for the treatment of cisplatin-induced acute kidney injury remains unclear. PURPOSE The objective of this study is to examine the impact of the YQJPXY on the inhibition of ferroptosis in cisplatin-induced acute kidney injury and to elucidate the underlying mechanisms. METHODS The active components of YQJPXY were analysed using UPLC-MS/MS. A comprehensive investigation was conducted to elucidate the effects and regulatory mechanisms of YQJPXY on CIAKI and ferroptosis in mice subjected to acute cisplatin treatment and in mice receiving cisplatin treatment after STING expression was inhibited using the STING inhibitor C176. The renoprotective effect of YQJPXY on cisplatin-treated mice was evaluated by measuring tissue damage, inflammation and pro-fibrosis. In addition, we employed network pharmacology and molecular docking methodologies to analyse the principal regulatory targets of YQJPXY. Furthermore, the expression of key proteins and markers of ferroptosis and iron metabolism, as well as the levels of key indicators related to STING-associated ferritinophagy, were examined by immunoblotting, immunohistochemistry, immunoprecipitation, quantitative real-time PCR (qPCR) and specific probes. RESULTS The results demonstrated that YQJPXY reduced the levels of indicators of injury, inflammation and pro-fibrosis in CIAKI mice, with renoprotective effects. Network pharmacological analyses revealed that ferroptosis might be the main biological process regulated by YQJPXY. Furthermore, molecular docking results indicated that STING might be a potential regulatory target of YQJPXY. Furthermore, YQJPXY treatment resulted in a significant reduction in MDA and 4-HNE levels, as well as the inhibition of ferroptosis and improvement in iron metabolic processes. Concomitantly, YQJPXY exhibited a robust protective effect on ferroptosis and iron metabolism homeostasis, as evidenced by its inhibitory action on ferritinophagy. Validation experiments utilising the cisplatin inhibitor C176 demonstrated that YQJPXY inhibits cisplatin-induced ferroptosis in kidney via STING-mediated ferritinophagy. CONCLUSION These suggest that YQJPXY alleviates cisplatin-induced acute kidney injury through suppressing ferroptosis via STING-NCOA4-mediated Ferritinophagy.
Collapse
Affiliation(s)
- Ji Zhu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou 330061, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Aini Yuan
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yifei Le
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaohui Chen
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Jianan Guo
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jing Liu
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hang Chen
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Cai-Yi Wang
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Dezhao Lu
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Keda Lu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou 330061, China.
| |
Collapse
|
36
|
He X, Zhong L, Wang N, Zhao B, Wang Y, Wu X, Zheng C, Ruan Y, Hou J, Luo Y, Yin Y, He Y, Xiang AP, Wang J. Gastric Cancer Actively Remodels Mechanical Microenvironment to Promote Chemotherapy Resistance via MSCs-Mediated Mitochondrial Transfer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404994. [PMID: 39392399 DOI: 10.1002/advs.202404994] [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: 05/08/2024] [Revised: 09/03/2024] [Indexed: 10/12/2024]
Abstract
Chemotherapy resistance is the main reason of treatment failure in gastric cancer (GC). However, the mechanism of oxaliplatin (OXA) resistance remains unclear. Here, we demonstrate that extracellular mechanical signaling plays crucial roles in OXA resistance within GC. We selected OXA-resistant GC patients and analyzed tumor tissues by single-cell sequencing, and found that the mitochondrial content of GC cells increased in a biosynthesis-independent manner. Moreover, we found that the increased mitochondria of GC cells were mainly derived from mesenchymal stromal cells (MSCs), which could repair the mitochondrial function and reduce the levels of mitophagy in GC cells, thus leading to OXA resistance. Furthermore, we investigated the underlying mechanism and found that mitochondrial transfer was mediated by mechanical signals of the extracellular matrix (ECM). After OXA administration, GC cells actively secreted ECM in the tumor microenvironment (TEM), increasing matrix stiffness of the tumor tissues, which promoted mitochondria to transfer from MSCs to GC cells via microvesicles (MVs). Meanwhile, inhibiting the mechanical-related RhoA/ROCK1 pathway could alleviate OXA resistance in GC cells. In summary, these results indicate that matrix stiffness could be used as an indicator to identify chemotherapy resistance, and targeting mechanical-related pathway could effectively alleviate OXA resistance and improve therapeutic efficacy.
Collapse
Affiliation(s)
- Xin He
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Li Zhong
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Nan Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Baiwei Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yannan Wang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinxiang Wu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Changyu Zheng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Yueheng Ruan
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Jianfeng Hou
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yusheng Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yuehan Yin
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yulong He
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiancheng Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
37
|
Barakat N, Ismail E, Zahran F. The integrated effect of roflumilast and selenium nanoparticles on nephrotoxicity generated by cisplatin through the regulation of the antioxidant and apoptotic pathways. J Trace Elem Med Biol 2024; 86:127555. [PMID: 39442470 DOI: 10.1016/j.jtemb.2024.127555] [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: 08/25/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
AIM The current investigations aimed to investigate the potential synergistic effect of Roflumilast (ROF) and Selenium nanoparticles (SeNPs) administration on Cisplatin (Cis) -induced nephrotoxicity. MATERIALS AND METHODS Fifty male rats were divided into five groups; Control group: animals were administered 0.9 % saline solution. Cis group: animals were injected with a single dose of 6 mg/kg. ROF group: Rats received a dosage of 1.2 mg/kg orally daily for 11 days. SeNPs group: animals orally received 0.5 mg/kg of ROF daily for 11 days. The ROF + SeNPs group was administered both after receiving a Cis injection for 11 days. Animals were sacrificed at 5 and 11 days, and the urine and blood samples were collected on day 5 and day 11 for chemical analysis, while kidney samples were obtained for molecular, histological, and immunohistochemical studies. RESULTS The levels of serum creatinine, Blood urea nitrogen (BUN), and total protein were elevated in the Cis group compared to the control group (p < 0.05). While the combination of ROF and SeNPs dramatically decreased these values after 5 and 11 days (p < 0.05). In addition, Cis caused renal oxidative stress by elevating MDA levels and suppressing the activities of SOD, GSH, and CAT. Similarly, these effects were modulated by ROF and SeNPs after 11 days (p < 0.05). Furthermore, the concurrent administration of ROF and SeNPs resulted in a significant increase in the expression of HO-1, Nrf2, and Bcl2, while decreasing the expression of BAX and IL-6 compared to the Cis group after 11 days (P < 0.05). CONCLUSION The study showed that both ROF and SeNPs had significant therapeutic potential in reducing the pathological alterations caused by Cis.
Collapse
Affiliation(s)
- Nashwa Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt.
| | - Ehab Ismail
- Biochemistry Division, Chemistry Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Faten Zahran
- Biochemistry Division, Chemistry Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| |
Collapse
|
38
|
Park J, Sim J, Yi HJ, Rhee SG, Woo HA. Cisplatin induces kidney damage through the down-regulation of Prx I by autophagic degradation. Free Radic Biol Med 2024; 225:236-246. [PMID: 39366472 DOI: 10.1016/j.freeradbiomed.2024.09.049] [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: 08/12/2024] [Revised: 09/15/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
In this study, we investigated the potential role of PrxI in cis-diamminedichloroplatinum (cisplatin)-induced renal damage in mice. The anticancer drug cisplatin is a chemotherapeutic agent that is widely used to treat solid tumors. Cisplatin-induced nephrotoxicity is a serious dose-limiting side effect, primarily caused by oxidative stress. The oxidative stress further damages DNA, membranes, and mitochondria, and increases endoplasmic reticulum (ER) stress. Cisplatin produces reactive oxygen species (ROS) through Cytochrome P450 2E1 (CYP2E1) and localizes to the surface of the ER, where CYP2E1 is located. Among the six Prx isoforms, Prx I was selectively degraded in cisplatin-treated kidneys during severe renal function damage. Prx I degradation is blocked in mouse proximal tubular cells treated with 3-methyladenine, an autophagy inhibitor, and in MEF lacking ATG7. Moreover, increased ROS levels on the ER surface due to CYP2E1 overexpression further accelerated Prx I degradation. These results suggest that Prx I degradation is largely mediated through autophagy, which is promoted by cisplatin-induced ER stress. Ablation of Prx I exacerbated cisplatin-induced nephrotoxicity and significantly increased the abundance of oxidative stress, ER stress, and inflammatory markers in the kidney, indicating that Prx I plays a protective role against cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Jiyoung Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea; Fluorescence Core Imaging Center, Department of Life Science, Ewha Womans University, Seoul, South Korea.
| | - Juhyun Sim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea; National Forensic Service, 26460, 10 Ipchun-ro, Wonju, Gangwon-do, South Korea.
| | - Ho Jin Yi
- College of Pharmacy, Graduate School of Applied Science and Technology for Skin Health and Aesthetics, Ewha Womans University, Seoul, 120-750, South Korea.
| | - Sue Goo Rhee
- Biochemistry and Biophysics Center, NHLBI, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Hyun Ae Woo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea; College of Pharmacy, Graduate School of Applied Science and Technology for Skin Health and Aesthetics, Ewha Womans University, Seoul, 120-750, South Korea.
| |
Collapse
|
39
|
Teng H, Sun X, Eglitis R, Wang X, Zhang W, Wang H, Qu S, Yu Z, Liu S, Zhao Y. Chiisanoside from the Leaves of Acanthopanax sessiliflorus Can Resist Cisplatin-Induced Ototoxicity by Maintaining Cytoskeletal Homeostasis and Inhibiting Ferroptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25720-25742. [PMID: 39505327 DOI: 10.1021/acs.jafc.4c07994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Ototoxicity is a common side effect of cisplatin cancer treatment, potentially leading to hearing loss. This study demonstrated the significant protective activity of Acanthopanax sessiliflorus (A. sessiliflorus) leaves against cisplatin-induced ototoxicity (CIO), investigated the active compounds, and elucidated their mechanisms in countering CIO. UPLC-Q/TOF-MS analysis identified 79 compounds. Network pharmacology and activity screening determined that chiisanoside (CSS) plays a crucial role in combating CIO. Transcriptomics combined with network pharmacology analysis and experiments revealed that CSS activates the Dock1/PIP5K1A pathway to suppress the actin-severing protein gelsolin, protecting hair cells from cisplatin-induced cytoskeleton damage. CSS also activates the SLC7A11/GPX4 pathway via TGFBR2, reducing lipid peroxidation and intracellular iron accumulation to suppress cisplatin-induced ferroptosis. This study discovers that the major component CSS in A. sessiliflorus leaves reverses CIO by regulating actin homeostasis via Dock1 and inhibiting ferroptosis through TGFBR2, providing a theoretical basis for expanding CIO treatment targets and related drug development.
Collapse
Affiliation(s)
- Hongbo Teng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin Province 130118, China
- International Joint Laboratory for Development of Animal and Plant Resources for Food and Medicine, Changchun, Jilin Province 130118, China
| | - Xialin Sun
- International Joint Laboratory for Development of Animal and Plant Resources for Food and Medicine, Changchun, Jilin Province 130118, China
- College of Pharmacy, Jilin Medical University, Jilin, Jilin Province 132013, China
| | - Roberts Eglitis
- Institute of Solid State Physics, University of Latvia, Riga LV-1067, Latvia
| | - Xv Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin Province 130118, China
- International Joint Laboratory for Development of Animal and Plant Resources for Food and Medicine, Changchun, Jilin Province 130118, China
| | - Wenxin Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin Province 130118, China
- International Joint Laboratory for Development of Animal and Plant Resources for Food and Medicine, Changchun, Jilin Province 130118, China
| | - Haijing Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin Province 130118, China
- International Joint Laboratory for Development of Animal and Plant Resources for Food and Medicine, Changchun, Jilin Province 130118, China
| | - Shurong Qu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin Province 130118, China
- International Joint Laboratory for Development of Animal and Plant Resources for Food and Medicine, Changchun, Jilin Province 130118, China
| | - Zhengxuan Yu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin Province 130118, China
| | - Shuangli Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin Province 130118, China
- International Joint Laboratory for Development of Animal and Plant Resources for Food and Medicine, Changchun, Jilin Province 130118, China
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin Province 130118, China
- International Joint Laboratory for Development of Animal and Plant Resources for Food and Medicine, Changchun, Jilin Province 130118, China
| |
Collapse
|
40
|
Balsa LM, Santa Maria de la Parra L, Ferretti V, León IE. Deciphering the Effect of a Cu(II)-hydrazone Complex on Intracellular Cell Signalling Pathways in a Human Osteosarcoma 2D and 3D Models. Chembiochem 2024; 25:e202400373. [PMID: 39121373 DOI: 10.1002/cbic.202400373] [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: 04/22/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/11/2024]
Abstract
New therapeutic strategies for osteosarcoma (OS) have demonstrated the potential efficacy of copper compounds as anticancer drugs and as a substitute for the often used platinum compounds. OS is a type of bone cancer, primarily affecting young adults and children.The main objective of this work is to discover the molecular targets and cellular pathways related to the antitumor properties of a Cu(II)-hydrazone toward human OS 2D and 3D systems. Cell viability study using MG-63 cells was evaluated in OS monolayer and spheroids. CuHL significantly reduced cell viability in OS models (IC50 2D: 2.6±0.3 μM; IC50 3D: 9.9±1.4 μM) (p<0.001). Also, CuHL inhibits cell proliferation and it induces cells to apoptosis. The main mechanism of action found for CuHL are the interaction with DNA, genotoxicity, the ROS generation and the proteasome activity inhibition. Besides, 67 differentially expressed proteins were found using proteomic approaches. Of those 67 proteins, 40 were found overexpressed and 27 underexpressed. The response to stress and to unfolded protein, as well as ATP synthesis were the most affected biological process among upregulated proteins, whilst proteins related to DNA replication and redox homeostasis were downregulated.
Collapse
Affiliation(s)
- Lucía M Balsa
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, (1900), Argentina
| | - Lucía Santa Maria de la Parra
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, (1900), Argentina
| | - Valeria Ferretti
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, (1900), Argentina
| | - Ignacio E León
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, (1900), Argentina
- Cátedra de Fisiopatología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, 1900), Argentina
| |
Collapse
|
41
|
Jeong M, Kurihara S, Stankovic KM. An In Vitro Oxidative Stress Model of the Human Inner Ear Using Human-Induced Pluripotent Stem Cell-Derived Otic Progenitor Cells. Antioxidants (Basel) 2024; 13:1407. [PMID: 39594548 PMCID: PMC11591063 DOI: 10.3390/antiox13111407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/30/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The inner ear organs responsible for hearing (cochlea) and balance (vestibular system) are susceptible to oxidative stress due to the high metabolic demands of their sensorineural cells. Oxidative stress-induced damage to these cells can cause hearing loss or vestibular dysfunction, yet the precise mechanisms remain unclear due to the limitations of animal models and challenges of obtaining living human inner ear tissue. Therefore, we developed an in vitro oxidative stress model of the pre-natal human inner ear using otic progenitor cells (OPCs) derived from human-induced pluripotent stem cells (hiPSCs). OPCs, hiPSCs, and HeLa cells were exposed to hydrogen peroxide or ototoxic drugs (gentamicin and cisplatin) that induce oxidative stress to evaluate subsequent cell viability, cell death, reactive oxygen species (ROS) production, mitochondrial activity, and apoptosis (caspase 3/7 activity). Dose-dependent reductions in OPC cell viability were observed post-exposure, demonstrating their vulnerability to oxidative stress. Notably, gentamicin exposure induced ROS production and cell death in OPCs, but not hiPSCs or HeLa cells. This OPC-based human model effectively simulates oxidative stress conditions in the human inner ear and may be useful for modeling the impact of ototoxicity during early pregnancy or evaluating therapies to prevent cytotoxicity.
Collapse
Affiliation(s)
- Minjin Jeong
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (S.K.)
| | - Sho Kurihara
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (S.K.)
- Department of Otolaryngology-Head and Neck Surgery, The Jikei University School of Medicine, 3-25-8 Nishishimbashi Minato-ku, Tokyo 105-8461, Japan
| | - Konstantina M. Stankovic
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (S.K.)
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
42
|
Ye M, Liu T, Miao L, Ji H, Xu Z, Wang H, Zhang J, Zhu X. Cisplatin-encapsulated TRAIL-engineered exosomes from human chorion-derived MSCs for targeted cervical cancer therapy. Stem Cell Res Ther 2024; 15:396. [PMID: 39497209 PMCID: PMC11536590 DOI: 10.1186/s13287-024-04006-6] [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: 06/24/2024] [Accepted: 10/21/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND Cisplatin (DDP) is an efficacious and widely applied chemotherapeutic drug for cervical cancer patients who are diagnosed as metastatic and inoperable, or desiring fertility preservation. Tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) selectively triggers cancer cells apoptosis by binding to cognate death receptors (DR4 and DR5). Mesenchymal stem cells-derived exosomes (MSCs-Exo) have been regarded as ideal drug carriers on account of their nanoscale, low toxicity, low immunogenicity, high stability, biodegradability, and abundant sources. METHODS Human chorion-derived mesenchymal stem cells (hCD-MSCs) were isolated by adherent culture method. TRAIL-engineered hCD-MSCs (hCD-MSCsTRAIL) were constructed by lentivirus transfection, and its secreted Exo (hCD-MSCs-ExoTRAIL) were acquired by differential centrifugation and confirmed to overexpress TRAIL by western blotting. Next, nanoscale drug delivery systems (DDP & hCD-MSCs-ExoTRAIL) were fabricated by loading DDP into hCD-MSCs-ExoTRAIL via electroporation. The CCK-8 assay and flow cytometry were conducted to explore the proliferation and apoptosis of cervical cancer cells (SiHa and HeLa), respectively. Cervical cancer-bearing nude mice were constructed to examine the antitumor activity and biosafety of DDP & hCD-MSCs-ExoTRAIL in vivo. RESULTS Compared with hCD-MSCs-Exo, hCD-MSCs-ExoTRAIL weakened proliferation and enhanced apoptosis of cervical cancer cells. DDP & hCD-MSCs-ExoTRAIL were proved to retard cervical cancer cell proliferation and propel cell apoptosis more effectively than DDP or hCD-MSCs-ExoTRAIL alone in vitro. In cervical cancer-bearing mice, DDP & hCD-MSCs-ExoTRAIL evidently hampered tumor growth, and its role in inducing apoptosis was mechanistically associated with JNK/p-c-Jun activation and survivin suppression. Moreover, DDP & hCD-MSCs-ExoTRAIL showed favorable biosafety in vivo. CONCLUSIONS DDP & hCD-MSCs-ExoTRAIL nanoparticles exhibited great promise for cervical cancer treatment as an Exo-based chemo-gene combinational therapy in clinical practice.
Collapse
Affiliation(s)
- Miaomiao Ye
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Tingxian Liu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Liqing Miao
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Huihui Ji
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Zhihui Xu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Huihui Wang
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Jian'an Zhang
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China.
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
43
|
Nguyen JP, Woerner LC, Johnson DE, Grandis JR. Future investigative directions for novel therapeutic targets in head and neck cancer. Expert Rev Anticancer Ther 2024; 24:1067-1084. [PMID: 39412140 PMCID: PMC11514385 DOI: 10.1080/14737140.2024.2417038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
AREAS COVERED Here we describe novel agents, their mechanism(s) of action, preclinical results, and ongoing clinical trials in HNSCC. EXPERT OPINION Established therapeutic targets in HNSCC include EGFR (cetuximab) and PD-1 (pembrolizumab and nivolumab). Despite the detection of many other possible targets in HNSCC cell lines and patient tumors, no other therapies have successfully advanced to date. Identification of predictive biomarkers may guide the use of targeted agents and combination therapies. Clinical trials supported by strong preclinical data in relevant models are more likely to advance treatment options.
Collapse
Affiliation(s)
- Jacqueline P. Nguyen
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, USA
| | - Liam C. Woerner
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, USA
| | - Daniel E. Johnson
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, USA
| | - Jennifer R. Grandis
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, USA
| |
Collapse
|
44
|
Pigg HC, Alley KR, Griffin CR, Moon CH, Kraske SJ, DeRose VJ. The unique Pt(II)-induced nucleolar stress response and its deviation from DNA damage response pathways. J Biol Chem 2024; 300:107858. [PMID: 39374783 PMCID: PMC11612370 DOI: 10.1016/j.jbc.2024.107858] [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: 06/05/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 10/09/2024] Open
Abstract
The mechanisms of action for the platinum compounds cisplatin and oxaliplatin have yet to be fully elucidated, despite the worldwide use of these drugs. Recent studies suggest that the two compounds may be working through different mechanisms, with cisplatin inducing cell death via the DNA damage response (DDR) and oxaliplatin utilizing a nucleolar stress-based cell death pathway. While cisplatin-induced DDR has been subject to much research, the mechanisms for oxaliplatin's influence on the nucleolus are not well understood. Prior work has outlined structural parameters for Pt(II) derivatives capable of nucleolar stress induction. In this work, we gain insight into the nucleolar stress response induced by these Pt(II) derivatives by investigating potential correlations between this unique pathway and DDR. Key findings from this study indicate that Pt(II)-induced nucleolar stress occurs when DDR is inhibited and works independently of the ATM/ATR-dependent DDR pathway. We also determine that Pt(II)-induced stress may be linked to the G1 cell cycle phase, as cisplatin can induce nucleolar stress when cell cycle inhibition occurs at the G1/S checkpoint. Finally, we compare Pt(II)-induced nucleolar stress with other small-molecule nucleolar stress-inducing compounds Actinomycin D, BMH-21, and CX-5461 and find that Pt(II) compounds cause irreversible nucleolar stress, whereas the reversibility of nucleolar stress induced by small-molecules varies. Taken together, these findings contribute to a better understanding of Pt(II)-induced nucleolar stress, its deviation from ATM/ATR-dependent DDR, and the possible influence of cell cycle on the ability of Pt(II) compounds to cause nucleolar stress.
Collapse
Affiliation(s)
- Hannah C Pigg
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | - Katelyn R Alley
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | | | - Caleb H Moon
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | - Sarah J Kraske
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | - Victoria J DeRose
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA.
| |
Collapse
|
45
|
Wu X, Peng X, Zhang Y, Peng W, Lu X, Deng T, Nie G. New application of ombuoside in protecting auditory cells from cisplatin-induced ototoxicity via the apoptosis pathway. Heliyon 2024; 10:e39166. [PMID: 39640804 PMCID: PMC11620119 DOI: 10.1016/j.heliyon.2024.e39166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
Hearing loss is caused by many factors including ototoxic drug-induced hair cell damage. Ombuoside, an antioxidant isolated from Gynostemma pentaphyllum, has been suggested to serve as a new neuroprotective drug. However, the role of ombuoside in protecting inner ear hair cells from ototoxic drug-induced damage has not been investigated. Here, we demonstrated the protective potential of ombuoside in mitigating drug-induced ototoxicity in vivo and in vitro. We used cisplatin, a highly ototoxic anti-tumor drug, to induce hair cell damage. Our results showed that ombuoside significantly increased the survival of cisplatin-treated HEI-OC1 cells. Further mechanism research suggested that ombuoside protects HEI-OCI cells from cisplatin-induced apoptosis by reducing the cisplatin-induced upregulation of apoptosis-promoting proteins Bax, Bak, as well as apoptosis indicator proteins cytochrome C and cleaved-caspase-3, and the downregulation of apoptosis-inhibiting proteins Bcl-2. Ombuoside also protects the cells from the excessive ROS production and mitochondrial membrane depolarization triggered by cisplatin. These results demonstrated the potential for ombuoside in protecting hair cells from cisplatin by suppressing ROS generation and the mitochondrial apoptotic cascade. Ombuoside showed promise in protecting hair cells from cisplatin-induced apoptosis by suppressing ROS generation and the mitochondrial apoptotic cascade. Furthermore, ombuoside co-treatment in mouse cochlear explants and zebrafish lateral neuromasts rescued the decreased number and deformed morphology of hair cells resulting from cisplatin exposure. These findings further validated our conclusions and indicated that ombuoside is a potential protector against hearing loss caused by ototoxicity as a clinical side effect of cisplatin.
Collapse
Affiliation(s)
- Xingxing Wu
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518035, China
| | - Xixia Peng
- Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yue Zhang
- Department of Otolaryngology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Wanjun Peng
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518035, China
| | - Xiaochan Lu
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Tingting Deng
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518035, China
| | - Guohui Nie
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518035, China
| |
Collapse
|
46
|
Zhu C, Li J, Sun W, Li D, Wang Y, Shen XC. Signaling Mechanism of Cuproptosis Activating cGAS-STING Immune Pathway. JACS AU 2024; 4:3988-3999. [PMID: 39483232 PMCID: PMC11522904 DOI: 10.1021/jacsau.4c00712] [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: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 11/03/2024]
Abstract
Copper-mediated programmed cell death, which influences the regulation of tumor progression, is an effective approach for antitumor molecular therapy. Unlike apoptosis, copper complex-induced cuproptosis by lipid-acylated protein aggregation triggers the mitochondrial proteotoxic stress response, which could be associated with immunomodulation. However, it remains a great challenge to understand the distinctive molecular mechanisms that presumably activate immunity by cuproptosis. Here, the new nonlabeling fluorescent molecular tools of Cu-DPPZ-Py+ and Cu-DPPZ-Ph are synthesized and used to investigate the differential immune signaling mechanisms induced by copper-mediated cuproptosis or apoptosis. With Cu-DPPZ-Py+ and Cu-Elesclomol, there is strong evidence that the triggering cuproptosis significantly drives mitochondrial DNA (mtDNA) release to activate innate immunity via cyclic GMP-AMP synthase-stimulation of interferon genes (cGAS-STING), which can improve T cell antitumor immunity in vivo. By contrast, it is observed that Cu-DPPZ-Ph treated tumor cells could release intracellular caspase-3, resulting in apoptosis-associated immunosuppression. This study supports insights into how cuproptosis bridges cGAS-STING immune pathways, contributing to the development of cuproptosis-based antitumor immunotherapy.
Collapse
Affiliation(s)
- Chengyuan Zhu
- State Key Laboratory for
Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory
for Chemistry and Molecular Engineering of Medicinal Resources (Ministry
of Education of China), Collaborative Innovation Center for Guangxi
Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Jialiang Li
- State Key Laboratory for
Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory
for Chemistry and Molecular Engineering of Medicinal Resources (Ministry
of Education of China), Collaborative Innovation Center for Guangxi
Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Wanying Sun
- State Key Laboratory for
Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory
for Chemistry and Molecular Engineering of Medicinal Resources (Ministry
of Education of China), Collaborative Innovation Center for Guangxi
Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Desheng Li
- State Key Laboratory for
Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory
for Chemistry and Molecular Engineering of Medicinal Resources (Ministry
of Education of China), Collaborative Innovation Center for Guangxi
Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yiliang Wang
- State Key Laboratory for
Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory
for Chemistry and Molecular Engineering of Medicinal Resources (Ministry
of Education of China), Collaborative Innovation Center for Guangxi
Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Xing-Can Shen
- State Key Laboratory for
Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory
for Chemistry and Molecular Engineering of Medicinal Resources (Ministry
of Education of China), Collaborative Innovation Center for Guangxi
Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
47
|
Slika A, Haydar C, Chacra JB, Al Alam S, Mehanna S, Lteif A, Elias MG, Deo KM, Taleb RI, Aldrich-Wright JR, Daher CF. Unveiling the chemotherapeutic potential of two platinum(IV) complexes in skin cancer: in vitro and in vivo Insights. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 7:100205. [PMID: 39554887 PMCID: PMC11566320 DOI: 10.1016/j.crphar.2024.100205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/30/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
The present study investigates the chemotherapeutic potential of two platinum (IV) complexes, P-PENT and P-HEX, against skin cancer in vitro and in vivo. Both complexes exhibited potent cytotoxicity against HaCaT-II-4 cells with IC50 values of 0.8 ± 0.08 μM and 1.3 ± 0.16 μM respectively, while demonstrating 8-10-fold selectivity compared to mesenchymal stem cells (MSCs). Western blot analysis revealed significant modulation of key apoptotic and survival pathways, including upregulation of Bax/Bcl2 ratio, cleaved caspase 3, and cytochrome c, suggesting induction of intrinsic apoptosis. The complexes also inhibited PI3K and MAPK pathways, as evidenced by decreased p-AKT/AKT and p-ERK/ERK ratios. Flow cytometry confirmed significant apoptotic cell death. Both complexes also increased reactive oxygen species production. In a DMBA/TPA-induced skin carcinogenesis mouse model, both complexes significantly suppressed tumor growth at doses considerably lower than the maximum tolerated dose, with no detectable toxicity. A dose escalation study in BALB/c mice showed that P-PENT and P-HEX were approximately 5-fold and 4-fold more tolerated than cisplatin, respectively. In conclusion, the present study provides evidence that P-PENT and P-HEX may have the characteristics of an effective and potentially safe anti-tumor drug that could be used in skin cancer treatment.
Collapse
Affiliation(s)
- Amjad Slika
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos, Mount Lebanon, Lebanon
| | - Christina Haydar
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos, Mount Lebanon, Lebanon
| | - Joelle Bou Chacra
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos, Mount Lebanon, Lebanon
| | - Seba Al Alam
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos, Mount Lebanon, Lebanon
| | - Stephanie Mehanna
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos, Mount Lebanon, Lebanon
| | - Anthony Lteif
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos, Mount Lebanon, Lebanon
| | - Maria George Elias
- School of Science, Western Sydney University, Locked Bag 1797 Penrith South, 2751, NSW, Australia
| | - Krishant M. Deo
- School of Science, Western Sydney University, Locked Bag 1797 Penrith South, 2751, NSW, Australia
| | - Robin I. Taleb
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos, Mount Lebanon, Lebanon
| | - Janice R. Aldrich-Wright
- School of Science, Western Sydney University, Locked Bag 1797 Penrith South, 2751, NSW, Australia
| | - Costantine F. Daher
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos, Mount Lebanon, Lebanon
- Alice Ramez Chagoury School of Nursing, Lebanese American University, Byblos, Mount Lebanon, Lebanon
| |
Collapse
|
48
|
Lv J, Chen J, Song Y, Yao Y, Wu G, Yuan D, Gu X, Li X, Xu C, Zhou B, Ye M, Lv T, Wang D, Song Y. Co-Delivery of VEGF siRNA and THPP via Metal-Organic Framework Reverses Cisplatin-Resistant Non-Small Cell Lung Cancer and Inhibits Metastasis through a MUC4 Regulating Mechanism. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56910-56925. [PMID: 39397733 DOI: 10.1021/acsami.4c15175] [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: 10/15/2024]
Abstract
Cisplatin resistance significantly impacts the antitumor efficacy of cisplatin chemotherapy and contributes to poor prognosis, including metastasis. In this study, we present the utilization of metal-organic framework (MOF) nanoparticles as the therapeutic component and drug loading scaffold for implementing a ternary combination therapeutic strategy to combat cisplatin-resistant lung cancer and metastasis. Specifically, by engineering MOFs (Cis@MOF-siVEGF) through the self-assembly of THPP as photosensitizer for photodynamic therapy (PDT), along with the incorporation of cisplatin (DDP) and VEGF siRNA (siVEGF), we propose the leverage of photodynamic-induced oxidative damage and gene silencing of the angiogenic factor to reverse cisplatin resistance and sensitize therapeutic potency. Our findings demonstrated that the chemo/photodynamic/antiangiogenic triple combination therapy via Cis@MOF-siVEGF under irradiation effectively inhibits cisplatin-resistant tumor growth and induces abscopal effects. Importantly, molecular mechanistic exploration suggested that MUC4 exerted regulatory effects on governing cancer metastasis, thus representing a potential immunotherapeutic target for cancer intervention. Overall, our study creates a MOFs-based multicomponent delivery platform for complementary therapeutic modules with synergistically enhanced antitumor efficacy and sheds light on potential regulatory mechanisms on cisplatin-resistance cancers.
Collapse
Affiliation(s)
- Jiawen Lv
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Jiayan Chen
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Yueyue Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing 210002, People's Republic of China
| | - Yanwen Yao
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Guannan Wu
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Dongmei Yuan
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Xiaoling Gu
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Xing Li
- Department of Endocrinology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Chunwei Xu
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, People's Republic of China
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Tangfeng Lv
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing 210002, People's Republic of China
| | - Dong Wang
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing 210002, People's Republic of China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing 210002, People's Republic of China
| |
Collapse
|
49
|
Cederroth CR, Dyhrfjeld-Johnsen J, Canlon B. Pharmacological Approaches to Hearing Loss. Pharmacol Rev 2024; 76:1063-1088. [PMID: 39164117 PMCID: PMC11549935 DOI: 10.1124/pharmrev.124.001195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024] Open
Abstract
Hearing disorders pose significant challenges to individuals experiencing them and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Current treatment options often focus on amplification devices, cochlear implants, or other rehabilitative therapies, leaving a substantial gap regarding effective pharmacological interventions. Advancements in our understanding of the molecular and cellular mechanisms involved in hearing disorders induced by noise, aging, and ototoxicity have opened new avenues for drug development, some of which have led to numerous clinical trials, with promising results. The development of optimal drug delivery solutions in animals and humans can also enhance the targeted delivery of medications to the ear. Moreover, large genome studies contributing to a genetic understanding of hearing loss in humans combined with advanced molecular technologies in animal studies have shown a great potential to increase our understanding of the etiologies of hearing loss. The auditory system exhibits circadian rhythms and temporal variations in its physiology, its vulnerability to auditory insults, and its responsiveness to drug treatments. The cochlear clock rhythms are under the control of the glucocorticoid system, and preclinical evidence suggests that the risk/benefit profile of hearing disorder treatments using chronopharmacological approaches would be beneficial. If translatable to the bedside, such approaches may improve the outcome of clinical trials. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug formulation and delivery as well as optimized timing of drug administration, holds great promise of more effective treatments. SIGNIFICANCE STATEMENT: Hearing disorders pose significant challenges to individuals and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug delivery procedures and optimized timing of drug administration, holds the promise of more effective treatments.
Collapse
Affiliation(s)
- Christopher R Cederroth
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Jonas Dyhrfjeld-Johnsen
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| |
Collapse
|
50
|
Jiang W, Wang G, Bai F, Hu B, Xu Y, Xu X, Nie G, Zhu WG, Chen F, Pei XH. BRCA1 Promotes Repair of DNA Damage in Cochlear Hair Cells and Prevents Hearing Loss. J Neurosci 2024; 44:e0132242024. [PMID: 39227158 PMCID: PMC11484548 DOI: 10.1523/jneurosci.0132-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024] Open
Abstract
Cochlear hair cells (HCs) sense sound waves and allow us to hear. Loss of HCs will cause irreversible sensorineural hearing loss. It is well known that DNA damage repair plays a critical role in protecting cells in many organs. However, how HCs respond to DNA damage and how defective DNA damage repair contributes to hearing loss remain elusive. In this study, we showed that cisplatin induced DNA damage in outer hair cells (OHCs) and promoted OHC loss, leading to hearing loss in mice of either sex. Cisplatin induced the expression of Brca1, a DNA damage repair factor, in OHCs. Deficiency of Brca1 induced OHC and hearing loss, and further promoted cisplatin-induced DNA damage in OHCs, accelerating OHC loss. This study provides the first in vivo evidence demonstrating that cisplatin mainly induces DNA damage in OHCs and that BRCA1 promotes repair of DNA damage in OHCs and prevents hearing loss. Our findings not only demonstrate that DNA damage-inducing agent generates DNA damage in postmitotic HCs but also suggest that DNA repair factors, like BRCA1, protect postmitotic HCs from DNA damage-induced cell death and hearing loss.
Collapse
Affiliation(s)
- Weitao Jiang
- International Cancer Center, Marshall Laboratory of Biomedical Engineering, Department of Otolaryngology, The First Affiliated Hospital, Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Guanrun Wang
- International Cancer Center, Marshall Laboratory of Biomedical Engineering, Department of Otolaryngology, The First Affiliated Hospital, Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Feng Bai
- Department of Pathology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Bing Hu
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Yang Xu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xingzhi Xu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and International Cancer Center, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Guohui Nie
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Medical School, Shenzhen 518060, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xin-Hai Pei
- International Cancer Center, Marshall Laboratory of Biomedical Engineering, Department of Otolaryngology, The First Affiliated Hospital, Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518060, China
| |
Collapse
|