1
|
Li Y, Zhang T, Mou Q, Liu S, Wu W, Wang S, Yan X, Liang J, Yan M, Liu W, Pan X. Overexpression of methionine sulfoxide reductase A alleviates acrylamide-induced neurotoxicity by mitigating lipid peroxidation and mitochondria-dependent apoptosis In vivo and In vitro. Food Chem Toxicol 2025; 199:115339. [PMID: 39986565 DOI: 10.1016/j.fct.2025.115339] [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: 12/15/2024] [Revised: 02/08/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Acrylamide (ACR) has garnered significant attention due to its neurotoxic effects. Oxidative stress, a key mechanism underlying ACR-induced neurotoxicity, is well-documented. Methionine sulfoxide reductase A (MsrA) plays a pivotal role in protecting various types of cells, including neuronal cells, against the effects of oxidative stress. However, the role of MsrA in ACR-induced neurotoxicity remains poorly understood. This study explored the effects of MsrA on ACR-induced neurotoxicity. After administering ACR by gavage at doses of 20 mg/kg, 30 mg/kg, and 40 mg/kg for 21 days, rats exhibited motor impairment and structural damage in the cerebellum. Both in vivo and in vitro, ACR dose-dependently reduced MsrA level, accompanied by increased reactive oxygen species (ROS) and malondialdehyde (MDA) levels, c-Jun N-terminal kinase (JNK) phosphorylation, and mitochondria-dependent neuronal apoptosis. To further ascertain the role of MsrA in mitigating ACR-induced neuronal apoptosis, SH-SY5Y cell line overexpressing MsrA was constructed. Overexpression of MsrA attenuated the ACR-induced increases in ROS and MDA levels. Additionally, alterations in mitochondrial membrane potential (MMP), mitochondrial ultrastructure, JNK phosphorylation, and mitochondria-dependent apoptosis caused by ACR were reversed in the cells overexpressing MsrA. These findings offer significant insights into the protective role of MsrA against ACR-induced neurotoxicity.
Collapse
Affiliation(s)
- Yuanyuan Li
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Tingting Zhang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Qiaoxing Mou
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Sirui Liu
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Wanxing Wu
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Simei Wang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xiaoyu Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Sichuan, 610075, China
| | - Jie Liang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Mengfan Yan
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Weiying Liu
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Xiaoqi Pan
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Sichuan, 610075, China.
| |
Collapse
|
2
|
Takács A, Jessen M, Lajkó E, Szász Z, Kalabay M, Csámpai A, Kőhidai L. Quinine-chalcone hybrids as potent inhibitors of P-glycoprotein with apoptotic effects on EBC-1 cells. Biomed Pharmacother 2025; 187:118076. [PMID: 40267640 DOI: 10.1016/j.biopha.2025.118076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025] Open
Abstract
Chincona alkaloids extracted from the bark stem of Cinchona officinalis have been historically used to treat fever and malaria. More recently, cinchona alkaloid derivatives have been attributed to apoptotic effects in the context of cancer. Similarly, chalcones are plant-derived polyphenolic compounds with known anti-fungal, -microbial, -malarial, and -carcinogenic properties. Here, we reveal cytotoxic and antiproliferative characteristics of synthetic quinine-chalcone hybrids in human cancer cell lines. Two derivatives (AD-12 and AD-13) presented IC50 values below 2 µM in the lung squamous cell carcinoma cell line (EBC-1). Our study shows that AD-12 and AD-13 increased intracellular ROS levels and promoted caspase-3/7, and -8 activity in EBC-1 cells. These apoptotic effects were accompanied by short-term inhibition of P-gp efflux activity, while expression levels of P-gp transporters remained stable. Together, our study illustrates the potential of quinine-chalcone hybrids as novel anticancer drug candidates.
Collapse
Affiliation(s)
- Angéla Takács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary.
| | - Malin Jessen
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary; Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Eszter Lajkó
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Zsófia Szász
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Márton Kalabay
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Antal Csámpai
- Institute of Chemistry, Eötvös Loránd University (ELTE), Budapest, Hungary
| | - László Kőhidai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
3
|
Liang Y, Lan H, Li Q, Gao M, Liu M, Xu Z, Gao Y, Zhang L, Li Y, Zhao B. Exploiting metabolic vulnerabilities through synergistic ferroptosis and disulfidptosis for breast cancer therapy. J Adv Res 2025:S2090-1232(25)00212-7. [PMID: 40164329 DOI: 10.1016/j.jare.2025.03.052] [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: 01/21/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025] Open
Abstract
INTRODUCTION Ferroptosis represents a promising therapeutic approach for breast cancer treatment. However, cancer cells can develop resistance through the SLC7A11-GSH-GPX4 axis, wherein increased SLC7A11 expression enhances cystine uptake, replenishes GSH, and reactivates GPX4. Notably, cells with high SLC7A11 expression become vulnerable to disulfidptosis under glucose-deprived conditions. OBJECTIVES We aimed to develop a dual-mode therapeutic strategy that simultaneously induces ferroptosis and disulfidptosis by targeting both lipid peroxidation and glucose metabolism in breast cancer cells. METHODS Fe-Cu-SS metal-organic frameworks (MOFs) loaded with BAY876 (FCSP@876 MOFs) were synthesized to enhance ferroptosis and trigger disulfidptosis in breast cancer cells. The MOFs were characterized using transmission electron microscopy (TEM), Fourier-transform infrared (FTIR) spectroscopy, X-ray photoelectron spectroscopy (XPS), and UV-Vis spectroscopy. In vitro experiments demonstrated that FCSP@876 MOFs increased reactive oxygen species (ROS) levels and lipid peroxidation while depleting NADPH. Western blotting and actin filament staining confirmed the underlying mechanisms. In vivo xenograft experiments in BALB/c mice assessed the synergistic effects of ferroptosis and disulfidptosis induction. RESULTS During ferroptosis induction, cancer cells exhibited an adaptive upregulation of SLC7A11 expression. FCSP@876 MOFs effectively counteracted this resistance mechanism by simultaneously inducing ferroptosis and restricting glucose uptake through BAY876, leading to NADPH depletion and subsequent disulfidptosis. Both in vitro and in vivo experiments demonstrated the enhanced therapeutic efficacy of this dual-mode strategy compared with single-mode treatments. CONCLUSION This study successfully developed a novel therapeutic strategy that combines ferroptosis and disulfidptosis using FCSP@876 MOFs, offering a promising approach for overcoming ferroptosis resistance in breast cancer therapy.
Collapse
Affiliation(s)
- Yu Liang
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haibo Lan
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qiuyu Li
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Mengdan Gao
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Minyi Liu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ziting Xu
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yang Gao
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Li Zhang
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yingjia Li
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Bingxia Zhao
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Experiment Education/Administration Center, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
4
|
Zhang J, Ali MY, Chong HB, Tien PC, Woods J, Noble C, Vornbäumen T, Ordulu Z, Possemato AP, Harry S, Fonticella JM, Fellah L, Harrison D, Ge M, Khandelwal N, Huang Y, Chauvin M, Bischof AT, Hambelton GM, Gohar MF, Zhang S, Choi M, Bouberhan S, Oliva E, Mino-Kenudson M, Pavlova NN, Lawrence M, Gainor JF, Beausoleil SA, Bardeesy N, Mostoslavsky R, Pépin D, Ott CJ, Liau B, Bar-Peled L. Oxidation of retromer complex controls mitochondrial translation. Nature 2025:10.1038/s41586-025-08756-y. [PMID: 40140582 DOI: 10.1038/s41586-025-08756-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/07/2025] [Indexed: 03/28/2025]
Abstract
Reactive oxygen species (ROS) underlie human pathologies including cancer and neurodegeneration1,2. However, the proteins that sense ROS levels and regulate their production through their cysteine residues remain ill defined. Here, using systematic base-editing and computational screens, we identify cysteines in VPS35, a member of the retromer trafficking complex3, that phenocopy inhibition of mitochondrial translation when mutated. We find that VPS35 underlies a reactive metabolite-sensing pathway that lowers mitochondrial translation to decrease ROS levels. Intracellular hydrogen peroxide oxidizes cysteine residues in VPS35, resulting in retromer dissociation from endosomal membranes and subsequent plasma membrane remodelling. We demonstrate that plasma membrane localization of the retromer substrate SLC7A1 is required to sustain mitochondrial translation. Furthermore, decreasing VPS35 levels or oxidation of its ROS-sensing cysteines confers resistance to ROS-generating chemotherapies, including cisplatin, in ovarian cancer models. Thus, we identify that intracellular ROS levels are communicated to the plasma membrane through VPS35 to regulate mitochondrial translation, connecting cytosolic ROS sensing to mitochondrial ROS production.
Collapse
Affiliation(s)
- Junbing Zhang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA.
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Md Yousuf Ali
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Harrison Byron Chong
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Pei-Chieh Tien
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - James Woods
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Carolina Noble
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Tristan Vornbäumen
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Zehra Ordulu
- Brigham and Women's Hospital, Department of Pathology, Harvard Medical School, MA, USA
| | | | - Stefan Harry
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Jay Miguel Fonticella
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Lina Fellah
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Drew Harrison
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Maolin Ge
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Neha Khandelwal
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Yingfei Huang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Maëva Chauvin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - Anica Tamara Bischof
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | | | - Magdy Farag Gohar
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Siwen Zhang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - MinGyu Choi
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sara Bouberhan
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Esther Oliva
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Natalya N Pavlova
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Michael Lawrence
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Nabeel Bardeesy
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Raul Mostoslavsky
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - Christopher J Ott
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Brian Liau
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Liron Bar-Peled
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA.
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Wang J, Shao F, Yu QX, Ye L, Wusiman D, Wu R, Tuo Z, Wang Z, Li D, Cho WC, Wei W, Feng D. The Common Hallmarks and Interconnected Pathways of Aging, Circadian Rhythms, and Cancer: Implications for Therapeutic Strategies. RESEARCH (WASHINGTON, D.C.) 2025; 8:0612. [PMID: 40046513 PMCID: PMC11880593 DOI: 10.34133/research.0612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/17/2025]
Abstract
The intricate relationship between cancer, circadian rhythms, and aging is increasingly recognized as a critical factor in understanding the mechanisms underlying tumorigenesis and cancer progression. Aging is a well-established primary risk factor for cancer, while disruptions in circadian rhythms are intricately associated with the tumorigenesis and progression of various tumors. Moreover, aging itself disrupts circadian rhythms, leading to physiological changes that may accelerate cancer development. Despite these connections, the specific interplay between these processes and their collective impact on cancer remains inadequately explored in the literature. In this review, we systematically explore the physiological mechanisms of circadian rhythms and their influence on cancer development. We discuss how core circadian genes impact tumor risk and prognosis, highlighting the shared hallmarks of cancer and aging such as genomic instability, cellular senescence, and chronic inflammation. Furthermore, we examine the interplay between circadian rhythms and aging, focusing on how this crosstalk contributes to tumorigenesis, tumor proliferation, and apoptosis, as well as the impact on cellular metabolism and genomic stability. By elucidating the common pathways linking aging, circadian rhythms, and cancer, this review provides new insights into the pathophysiology of cancer and identifies potential therapeutic strategies. We propose that targeting the circadian regulation of cancer hallmarks could pave the way for novel treatments, including chronotherapy and antiaging interventions, which may offer important benefits in the clinical management of cancer.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Fanglin Shao
- Department of Rehabilitation,
The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qing Xin Yu
- Department of Pathology,
Ningbo Clinical Pathology Diagnosis Center, Ningbo, Zhejiang 315211, China
- Department of Pathology,
Ningbo Medical Centre Lihuili Hospital, Ningbo, Zhejiang 315040, China
| | - Luxia Ye
- Department of Public Research Platform,
Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47906, USA
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Zhouting Tuo
- Department of Urological Surgery, Daping Hospital, Army Medical Center of PLA,
Army Medical University, Chongqing, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People’s Hospital,
University of Electronic Science and Technology of China, Chengdu, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - William C. Cho
- Department of Clinical Oncology,
Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
- Division of Surgery and Interventional Science,
University College London, London W1W 7TS, UK
| |
Collapse
|
6
|
Shang S, Zheng F, Tan W, Xing Z, Chen S, Peng F, Lv X, Wang D, Zhu X, Wu J, Zhou Z, Zhang X, Yang X. Piezoelectric Biomaterial with Advanced Design for Tissue Infection Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413105. [PMID: 39887897 PMCID: PMC11905007 DOI: 10.1002/advs.202413105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/25/2024] [Indexed: 02/01/2025]
Abstract
Bacterial infection has become the most dangerous factor in tissue repair, which strongly affects the tissue regeneration efficiency and wellness of patients. Piezoelectric materials exhibit the outstanding advantage of producing electrons without external power supply. The ability of electron enrichment and reactive oxygen species generation through noninvasive stimulations enables piezoelectric materials the potential applications of antibacterial. Many studies have proved the feasibility of piezoelectric materials as a functional addition in antibacterial biomaterial. In fact, numerous piezoelectric materials with ingenious designs are reported to be effective in antibacterial processes. This review summarizes the antibacterial mechanisms of piezoelectric, illuminating their potential in combating bacteria. Recent advancement in the design and construction of piezoelectric biomaterial including defect engineering, heterojunction, synergy with metal and the composite scaffold configuration are thoroughly reviewed. Moreover, the applications and therapeutic effects of piezoelectric materials in common tissues with antibacterial requirements are introduced, such as orthopedics, dental, and wound healing. Finally, the development prospects and points deserving further exploration are listed. This review is expected to provide valuable insight into the relationship between antibacterial processes and piezoelectric materials, further inspiring constructive development in this emerging scientific discipline.
Collapse
Affiliation(s)
- Siyuan Shang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Fuyuan Zheng
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, 610065, China
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wen Tan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Zhengyi Xing
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Siyu Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
- Department of Burn and Plastic Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fuli Peng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Xiang Lv
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Duan Wang
- Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, 610065, China
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Jiagang Wu
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610065, China
- College of Physics, Sichuan University, Chengdu, 610065, China
| | - Zongke Zhou
- Orthopedic Research Institute and Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
7
|
Lam W, Yao Y, Tang C, Wang Y, Yuan Q, Peng L. Bifunctional mesoporous HMUiO-66-NH 2 nanoparticles for bone remodeling and ROS scavenging in periodontitis therapy. Biomaterials 2025; 314:122872. [PMID: 39383779 DOI: 10.1016/j.biomaterials.2024.122872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/17/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Periodontal bone defects represent an irreversible consequence of periodontitis associated with reactive oxygen species (ROS). However, indiscriminate removal of ROS proves to be counterproductive for tissue repair and insufficient for addressing existing bone defects. In the treatment of periodontitis, it is crucial to rationally alleviate local ROS while simultaneously promoting bone regeneration. In this study, Zr-based large-pore hierarchical mesoporous metal-organic framework (MOF) nanoparticles (NPs) HMUiO-66-NH2 were successfully proposed as bifunctional nanomaterials for bone regeneration and ROS scavenging in periodontitis therapy. HMUiO-66-NH2 NPs demonstrated outstanding biocompatibility both in vitro and in vivo. Significantly, these NPs enhanced the osteogenic differentiation of bone mesenchymal stem cells (BMSCs) under normal and high ROS conditions, upregulating osteogenic gene expression and mitigating oxidative stress. Furthermore, in vivo imaging revealed a gradual degradation of HMUiO-66-NH2 NPs in periodontal tissues. Local injection of HMUiO-66-NH2 effectively reduced bone defects and ROS levels in periodontitis-induced C57BL/6 mice. RNA sequencing highlighted that differentially expressed genes (DEGs) are predominantly involved in bone tissue development, with notable upregulation in Wnt and TGF-β signaling pathways. In conclusion, HMUiO-66-NH2 exhibits dual functionality in alleviating oxidative stress and promoting bone repair, positioning it as an effective strategy against bone resorption in oxidative stress-related periodontitis.
Collapse
Affiliation(s)
- Waishan Lam
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yufei Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, China
| | - Chenxi Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yue Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Surgery, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lin Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
8
|
Yang G, Milne GL, Nogueira MS, Yi H, Lan Q, Gao YT, Shu XO, Zheng W, Chen Q. Lipid peroxidation and colorectal cancer risk: a time-varying relationship. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.16.25322362. [PMID: 40034784 PMCID: PMC11875262 DOI: 10.1101/2025.02.16.25322362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Importance We recently observed an inverse and time-dependent association between systemic oxidative stress (OxS), measured by urinary biomarkers of nucleic acid oxidation, and colorectal cancer (CRC) risk. Further investigations into other types of OxS markers are warranted. Objective To extend the investigation into systemic lipid peroxidation and CRC risk. Design Setting and Participants Utilizing a nested case-control design, this study's primary analysis was performed in two large prospective cohorts in Shanghai, China, and replicated in an independent cohort in the US. Exposures Systemic lipid peroxidation was assessed by urinary F 2 -isoprostanes (F 2 -IsoPs) using UPLC-MS/MS assays. Main Outcomes and Measures During 15.1-year follow-up in the Shanghai cohorts, 1938 incident CRC cases were identified and matched to one control each through incidence-density sampling. In the US cohort, 285 incident CRC cases were included, each matched to two controls. Odds ratios (ORs) for CRC were calculated using multivariable conditional logistic regression models. Results Elevated levels of urinary 5-F 2t -IsoP (5-iPF 2α -VI), a major isomer of F 2 -IsoPs induced solely by free radicals, were associated with reduced risk of CRC in the Shanghai cohorts. This finding was replicated in the US cohort. Moreover, this inverse association was time-dependent, manifesting only in the later years of cancer development. Multivariable-adjusted ORs (95% CI) for CRC diagnosed within 5 years of enrollment at the 10th and 90th percentiles of 5-F 2t -IsoP levels, relative to the median, were 1.57 (1.26-1.96) and 0.61 (0.42-0.89), respectively, indicating a 2.2-fold difference in risk between the two groups. A stronger association was observed when using the composite index of DNA, RNA and lipid OxS markers, showing a 3.9-fold difference in risk between the two groups. No significant association was found for CRC diagnosed beyond 5 years of enrollment. Conclusions This study provides new evidence that systemic OxS is inversely and time-dependently associated with CRC risk in humans. Key Points Question: Is the time-dependent relationship between oxidative stress and tumorigenesis observed at the cellular level in experimental models also present at the systemic level in a population-based setting?Findings: Elevated systemic lipid peroxidation, measured by urinary F 2 -isoprostanes, was associated with a reduced risk of colorectal cancer (CRC) in two large prospective cohort studies in Shanghai, China, which was replicated in an independent cohort in the United States. This association varied over time, showing a stronger effect as cancer advanced. Meaning: This study provides new evidence that systemic oxidative stress is inversely and time-dependently associated with CRC risk in humans.
Collapse
|
9
|
Ženíšková K, Stopka P, Martín-Pérez T, Chevreux G, Grechnikova M, Drncová E, Malych R, Mach J, Walochnik J, Camadro JM, Sutak R. Molecular Mechanisms of Acanthamoeba castellanii Response to Different Sources of Oxidative Stress. J Proteome Res 2025; 24:449-458. [PMID: 39829028 PMCID: PMC11812009 DOI: 10.1021/acs.jproteome.4c00573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/27/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
Oxidative stress is a biological principle affecting all life on Earth and is also an important factor in the pathogen-host relationship. The pathogenic free-living amoeba Acanthamoeba castellanii has several pathways to cope with reactive oxygen species and the damage that they cause. In this study, we aimed to provide a comprehensive analysis of the amoeba's response to different sources of oxidative stress. Using whole-cell proteomic analysis, we obtained a complex picture of the changes in the proteome and identified potential key players in the defense against oxidative stress. Importantly, from the differential proteomics analysis, we identified a candidate efflux pump that may be involved in Acanthamoeba drug resistance.
Collapse
Affiliation(s)
- Kateřina Ženíšková
- Department
of Parasitology, Faculty of Science, BIOCEV, Charles University, Vestec 25250, Czech Republic
| | - Pavel Stopka
- Department
of Zoology, Faculty of Science, BIOCEV, Charles University, Vestec 25250, Czech Republic
| | - Tania Martín-Pérez
- Center
for Pathophysiology, Infectiology and Immunology, Institute of Specific
Prophylaxis and Tropical Medicine, Medical
University of Vienna, Vienna 1090, Austria
| | - Guillaume Chevreux
- Université
de Paris Cité, CNRS, Institut Jacques Monod, Paris F-75013, France
| | - Maria Grechnikova
- Department
of Parasitology, Faculty of Science, BIOCEV, Charles University, Vestec 25250, Czech Republic
| | - Eliška Drncová
- Department
of Parasitology, Faculty of Science, BIOCEV, Charles University, Vestec 25250, Czech Republic
| | - Ronald Malych
- Department
of Parasitology, Faculty of Science, BIOCEV, Charles University, Vestec 25250, Czech Republic
| | - Jan Mach
- Department
of Parasitology, Faculty of Science, BIOCEV, Charles University, Vestec 25250, Czech Republic
| | - Julia Walochnik
- Center
for Pathophysiology, Infectiology and Immunology, Institute of Specific
Prophylaxis and Tropical Medicine, Medical
University of Vienna, Vienna 1090, Austria
| | - Jean-Michel Camadro
- Université
de Paris Cité, CNRS, Institut Jacques Monod, Paris F-75013, France
| | - Robert Sutak
- Department
of Parasitology, Faculty of Science, BIOCEV, Charles University, Vestec 25250, Czech Republic
| |
Collapse
|
10
|
Kramer‐Drauberg M, Petrini E, Mira A, Patrucco E, Scardaci R, Savinelli I, Wang H, Qiao K, Carrà G, Nokin M, Zhou Z, Westover KD, Santamaria D, Porporato PE, Ambrogio C. Oncogenic mutant KRAS inhibition through oxidation at cysteine 118. Mol Oncol 2025; 19:311-328. [PMID: 39838816 PMCID: PMC11793020 DOI: 10.1002/1878-0261.13798] [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: 06/09/2024] [Revised: 10/30/2024] [Accepted: 12/24/2024] [Indexed: 01/23/2025] Open
Abstract
Specific reactive oxygen species activate the GTPase Kirsten rat sarcoma virus (KRAS) by reacting with cysteine 118 (C118), leading to an electron transfer between C118 and nucleoside guanosine diphosphate (GDP), which causes the release of GDP. Here, we have mimicked permanent oxidation of human KRAS at C118 by replacing C118 with aspartic acid (C118D) in KRAS to show that oncogenic mutant KRAS is selectively inhibited via oxidation at C118, both in vitro and in vivo. Moreover, the combined treatment of hydrogen-peroxide-producing pro-oxidant paraquat and nitric-oxide-producing inhibitor N(ω)-nitro-l-arginine methyl ester selectively inhibits human mutant KRAS activity by inducing oxidization at C118. Our study shows for the first time the vulnerability of human mutant KRAS to oxidation, thereby paving the way to explore oxidation-based anti-KRAS treatments in humans.
Collapse
Affiliation(s)
- Maximilian Kramer‐Drauberg
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Ettore Petrini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Alessia Mira
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Enrico Patrucco
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Rossella Scardaci
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Ilenia Savinelli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Haiyun Wang
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Keying Qiao
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Giovanna Carrà
- Department of Clinical and Biological SciencesUniversity of TorinoOrbassanoItaly
| | - Marie‐Julie Nokin
- Laboratory of Tumor and Development Biology, GIGA‐CancerUniversity of LiegeBelgium
| | - Zhiwei Zhou
- Department of BiochemistryThe University of Texas Southwestern Medical CenterDallasTXUSA
- Department of Radiation OncologyThe University of Texas Southwestern Medical CenterDallasTXUSA
| | - Kenneth D. Westover
- Department of BiochemistryThe University of Texas Southwestern Medical CenterDallasTXUSA
- Department of Radiation OncologyThe University of Texas Southwestern Medical CenterDallasTXUSA
| | - David Santamaria
- Molecular Mechanisms of Cancer Program, Centro de Investigación del CáncerCSIC‐Universidad de SalamancaSpain
| | - Paolo E. Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| |
Collapse
|
11
|
Summer M, Riaz S, Ali S, Noor Q, Ashraf R, Khan RRM. Understanding the Dual Role of Macrophages in Tumor Growth and Therapy: A Mechanistic Review. Chem Biodivers 2025:e202402976. [PMID: 39869825 DOI: 10.1002/cbdv.202402976] [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: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
Macrophages are heterogeneous cells that are the mediators of tissue homeostasis. These immune cells originated from monocytes and are classified into two basic categories, M1 and M2 macrophages. M1 macrophages exhibit anti-tumorous inflammatory reactions due to the behavior of phagocytosis. M2 macrophages or tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) and have a basic role in tumor progression by interacting with other immune cells in TME. By the expression of various cytokines, chemokines, and growth factors, TAMs lead to strengthening tumor cell proliferation, angiogenesis, and suppression of the immune system which further support invasion and metastasis. This review discusses recent and updated mechanisms regarding tumor progression by M2 macrophages. Moreover, the current therapeutic approaches targeting TAMs, their advantages, and limitations are also summarized, and further treatment approaches are outlined along with an elaboration of the tumor regression role of macrophages. This comprehensive review article possibly helps to understand the mechanisms underlying the tumor progression and regression role of macrophages in a comparative way from a basic level to the advanced one.
Collapse
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Saima Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Qudsia Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rimsha Ashraf
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rana Rashad Mahmood Khan
- Faculty of Chemistry and Life Sciences, Department of Chemistry, Government College University Lahore, Lahore, Pakistan
| |
Collapse
|
12
|
Baruah P, Padhi D, Moorthy H, Ramesh M, Govindaraju T. Navigating the dichotomy of reactive oxygen, nitrogen, and sulfur species: detection strategies and therapeutic interventions. RSC Chem Biol 2025:d5cb00006h. [PMID: 39877134 PMCID: PMC11770382 DOI: 10.1039/d5cb00006h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Reactive oxygen, nitrogen and sulfur species (RONSS) collectively encompasses a variety of energetically dynamic entities that emerge as inherent characteristics of aerobic life. This broad category includes reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species (RSS). A conundrum arises from the indispensable role of RONSS in redox signalling, while its overproduction in the mitochondria poses deleterious effects. This imbalance leads to biomolecular damage and contributes to neurodegenerative disorders, cancer, cardiovascular diseases and inflammation. Notably, the differential roles of RONSS across various diseases can be strategically exploited for therapeutic interventions. Timely, precise, and sensitive detection methods are indispensable for elucidating the spatiotemporal dynamics of RONSS and evaluating disease pathogenesis and progression. By monitoring RONSS levels, we can discern early markers of disease onset, enabling proactive intervention strategies for effective disease management. Therapeutic interventions targeting oxidative/nitrosative stress in disease pathologies have proven to be effective treatment routes in the mitigation of different diseases. This review aims to offer a comprehensive overview of the functional implications and delicate balance of RONSS in disease conditions, and advances made in detection strategies over the years while offering therapeutic strategies to tackle their adverse effects. A special emphasis is focussed on neurodegenerative disorders and cancer with case studies using RONSS-targeted chemical probes and prodrugs.
Collapse
Affiliation(s)
- Prayasee Baruah
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| | - Dikshaa Padhi
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| | - Hariharan Moorthy
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| |
Collapse
|
13
|
Zhou S, Zhao Z, Wang Z, Xu H, Li Y, Xu K, Li W, Yang J. Cancer-associated fibroblasts in carcinogenesis. J Transl Med 2025; 23:50. [PMID: 39806363 PMCID: PMC11727299 DOI: 10.1186/s12967-025-06071-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
In contemporary times, cancer poses the most significant threat to human life and safety. Scientists have relentlessly pursued the intricacies of carcinogenesis and explored ways to prevent and treat cancer. Carcinogenesis is a complex, multi-faceted, and multi-stage process, with numerous underlying causes, including inflammation and fibrosis. Cancer-associated fibroblasts (CAFs), however, occupy a pivotal and substantial role within the tumor microenvironment, facilitating carcinogenesis through diverse mechanisms such as creating inflammation, fostering a fibrotic tumor microenvironment, and immunosuppression. In this paper, we introduce the concept of carcinogenesis, explain its causes, describe the characteristics of CAFs and their sources, and highlight the roles and mechanisms of CAFs in promoting carcinogenesis. Ultimately, our aim is to contribute to the development of novel therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Shufen Zhou
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zekun Zhao
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhaojun Wang
- Department of Thyroid and Breast Surgery, The DingLi Clinical, The Wenzhou Central Hospital, College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hanzheng Xu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yijie Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China.
| | - Wei Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Jiahua Yang
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|
14
|
Zheng C, Pan Y, Lin B, Li J, Chen Q, Zheng Z. NRF3 suppresses the metastasis of triple-negative breast cancer cells by inhibiting ERK activation in a ROS-dependent manner. Histol Histopathol 2025; 40:123-131. [PMID: 39005145 DOI: 10.14670/hh-18-786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
PURPOSE Our previous study demonstrated that NRF3 (NFE2L3, Nuclear Factor-erythroid 2-related factor 3) could suppress cell metastasis and proliferation in breast cancer. In this study, we investigated the mechanisms underlying its function in breast cancer. METHODS In the present study, NRF3 expression and its clinical characteristics in breast cancer were analyzed using public datasets and clinical specimens. After breast cancer cells were overexpressed NRF3, FACS was used to detect the intracellular ROS levels. The migration and invasion activities of NRF3-ectopic expressed breast cancer cells were determined by transwell assay. To validate the role of ROS/ERK axis in the inhibitory effect of NRF3 in cell metastasis, ROS scavenger NAC was also included. RESULTS We found that NRF3 mRNA was highly expressed, while NRF3 protein was extremely lowly expressed in breast cancer tissues compared with their normal counterparts, and low level NRF3 was associated with poorer prognosis in patients with triple negative breast cancer (TNBC). More interestingly, overexpression of NRF3 protein significantly increased cellular ROS production and dramatically decreased p-ERK level and cell migration in TNBC cells. Mechanistically, NRF3 protein was found to be mutually regulated by valosin-containing protein (VCP). Strikingly, VCP-knockdown dramatically increased NRF3 protein expression, but NRF3-knockin also decreased VCP expression in return. Moreover, antioxidant NAC treatment effectively increased the level of p-ERK and VCP expression, as well as cell migration and invasion abilities of TNBC cells. CONCLUSION NRF3, a tumor suppressor downregulated by VCP, could attenuate cell metastasis in TNBC cells by increasing cellular ROS accumulation and subsequently inhibiting the ERK phosphorylation.
Collapse
Affiliation(s)
- Chenhui Zheng
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital) Taizhou, PR China
| | - Yue Pan
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital) Taizhou, PR China
| | - Bangyi Lin
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital) Taizhou, PR China
| | - Jin Li
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University Hospital) Taizhou, PR China
| | - Qi Chen
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital) Taizhou, PR China
| | - Zhibao Zheng
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital) Taizhou, PR China.
| |
Collapse
|
15
|
Wei B, Yang Z, Guo H, Wang Y, Chen W, Zhou J, Jin R, Wang Z, Tang Y. Design, synthesis, and biological evaluation of evodiamine-indolequinone hybrids as novel NQO1 agonists against non-small cell lung cancer. ARAB J CHEM 2025; 18:106075. [DOI: 10.1016/j.arabjc.2024.106075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024] Open
|
16
|
Rizwan M, Mahjabeen I, Haris MS, Qayyum F, Kayani MA. Deregulation of Exosomal miR-17, miR-20a and TGFBR2 in Head and Neck Cancer Patients. Technol Cancer Res Treat 2025; 24:15330338251323314. [PMID: 39989256 PMCID: PMC11848883 DOI: 10.1177/15330338251323314] [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: 02/25/2025] Open
Abstract
Introduction: Exosomes play significant roles in transferring cargo materials like proteins, RNAs (including miRNAs), and DNA. However, the role of serum exosome shuttled RNAs and miRNAs in head and neck cancer (HNC) remains unclear. This study assessed the diagnostic and prognostic significance of exosomal miR-17, miR-20a, and TGFBR2 in HNC patients. Methods: Exosomes were isolated, from 400 confirmed HNC patients and 400 healthy controls, and characterized by NTA, TEM, Immunolabelling, and ELISA. Quantitative PCR was used to check the expressions of exosomal molecules. Oxidative stress was also measured through ELISA in cancer patients and healthy controls. Results: Data analysis revealed significant dysregulation in the expressional levels of miR-17 (p < .0001), miR-20a (p = .0003), and TGFBR2 (p = .0005), which were found associated with aggressiveness and poor survival of HNC patients. Spearman correlation revealed a positive statistically significant association between miR-20a versus miR-17 (r = 0.534; p < .01), while a negative correlation was found between TGFBR2 versus miR-17 (r = -0.240; p = .015). Significantly decreased levels of peroxidase (POD) (p < .0001) and an increased level of 8-Oxoguanine (p < .0001) were observed. Conclusion: The results showed that these exosomal miRNAs and target gene may serve as potential and noninvasive diagnostic and prognostic markers for head and neck cancer patients.
Collapse
Affiliation(s)
- Muhammad Rizwan
- Cancer Genetics & Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Ishrat Mahjabeen
- Cancer Genetics & Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Muhammad Shahbaz Haris
- Cancer Genetics & Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Fouzia Qayyum
- Department of ENT, Bahawalpur Victoria Hospital, Bahawalpur, Pakistan
| | - Mahmood Akhtar Kayani
- Cancer Genetics & Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
17
|
Wang J, Li C, Han J, Xue Y, Zheng X, Wang R, Radak Z, Nakabeppu Y, Boldogh I, Ba X. Reassessing the roles of oxidative DNA base lesion 8-oxoGua and repair enzyme OGG1 in tumorigenesis. J Biomed Sci 2025; 32:1. [PMID: 39741341 DOI: 10.1186/s12929-024-01093-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 11/08/2024] [Indexed: 01/02/2025] Open
Abstract
ROS cause multiple forms of DNA damage, and among them, 8-oxoguanine (8-oxoGua), an oxidized product of guanine, is one of the most abundant. If left unrepaired, 8-oxoGua may pair with A instead of C, leading to a mutation of G: C to T: A during DNA replication. 8-Oxoguanine DNA glycosylase 1 (OGG1) is a tailored repair enzyme that recognizes 8-oxoGua in DNA duplex and initiates the base excision repair (BER) pathway to remove the lesion and ensure the fidelity of the genome. The accumulation of genomic 8-oxoGua and the dysfunction of OGG1 is readily linked to mutagenesis, and subsequently aging-related diseases and tumorigenesis; however, the direct experimental evidence has long been lacking. Recently, a series of studies have shown that guanine oxidation in the genome has a conservative bias, with the tendency to occur in the regulatory regions, thus, 8-oxoGua is not only a lesion to be repaired, but also an epigenetic modification. In this regard, OGG1 is a specific reader of this base modification. Substrate recognition and/or excision by OGG1 can cause DNA conformation changes, affect chromatin modifications, thereby modulating the transcription of genes involved in a variety of cellular processes, including inflammation, cell proliferation, differentiation, and apoptosis. Thus, in addition to the potential mutagenicity, 8-oxoGua may contribute to tumor development and progression through the altered gene expression stemming from its epigenetic effects.
Collapse
Affiliation(s)
- Jing Wang
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130031, China
| | - Chunshuang Li
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Jinling Han
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Yaoyao Xue
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Xu Zheng
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Ruoxi Wang
- College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Zsolt Radak
- Research Institute of Sport Science, University of Physical Education, Budapest, 1123, Hungary
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA.
| | - Xueqing Ba
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, 130024, China.
| |
Collapse
|
18
|
Nath P, Alfarsi LH, El-Ansari R, Masisi BK, Erkan B, Fakroun A, Ellis IO, Rakha EA, Green AR. The amino acid transporter SLC7A11 expression in breast cancer. Cancer Biol Ther 2024; 25:2291855. [PMID: 38073087 PMCID: PMC10761065 DOI: 10.1080/15384047.2023.2291855] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer (BC), characterized by its diverse molecular profiles and clinical outcomes, presents a significant challenge in the development of effective therapeutic strategies. Metabolic reprogramming, a defining characteristic of cancer, has emerged as a promising target for novel therapies. SLC7A11, an amino acid transporter that facilitates cysteine uptake in exchange for glutamate, plays a crucial role in sustaining the altered metabolism of cancer cells. This study delves into the comprehensive analysis of SLC7A11 at the genomic, transcriptomic, and protein levels in extensive BC datasets to elucidate its potential role in different BC subtypes. SLC7A11 gene copy number and mRNA expression were evaluated using the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) cohort (n = 1,980) and Breast Cancer Gene Expression Miner (n = 4,712). SLC7A11 protein was assessed using immunohistochemistry in a large BC cohort (n = 1,981). Additionally, The Cancer Genome Atlas (TCGA) dataset was used to explore SLC7A11 DNA methylation patterns using MethSurv (n = 782) and association of SLC7A11 mRNA expression with immune infiltrates using TIMER (n = 1,100). High SLC7A11 mRNA and SLC7A11 protein expression were significantly associated with high tumor grade (p ≤ .02), indicating a potential role in cancer progression. Interestingly, SLC7A11 copy number gain was observed in HER2+ tumors (p = .01), suggesting a subtype-specific association. In contrast, SLC7A11 mRNA expression was higher in the basal-like/triple-negative (TN; p < .001) and luminal B tumors (p = .02), highlighting its differential expression across BC subtypes. Notably, high SLC7A11 protein expression was predominantly observed in Estrogen Receptor (ER)-negative and Triple Negative (TN) BC, suggesting a role in these aggressive subtypes. Further analysis revealed that SLC7A11 was positively correlated with other amino acid transporters and enzymes associated with glutamine metabolism, implying a coordinated role in metabolic regulation. Additionally, SLC7A11 gene expression was positively associated with neutrophil and macrophage infiltration, suggesting a potential link between SLC7A11 and tumor immunity. Our findings suggest that SLC7A11 plays a significant role in BC metabolism, demonstrating differential expression across subtypes and associations with poor patient outcomes. Further functional studies are warranted to elucidate the precise mechanisms by which SLC7A11 contributes to BC progression and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Preyanka Nath
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Lutfi H. Alfarsi
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Rokaya El-Ansari
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Brendah K. Masisi
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Busra Erkan
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Ali Fakroun
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Ian O. Ellis
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
- Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Emad A. Rakha
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
- Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Andrew R. Green
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| |
Collapse
|
19
|
Li Y, Zhang L, Xu G, Xu G, Chen J, Zhao K, Li M, Jin J, Peng C, Wang K, Pan S, Zhu K. Exploration and validation of a novel reactive oxygen species-related signature for predicting the prognosis and chemotherapy response of patients with bladder cancer. Front Immunol 2024; 15:1493528. [PMID: 39749345 PMCID: PMC11693660 DOI: 10.3389/fimmu.2024.1493528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Background Reactive Oxygen Species (ROS), a hallmark of cancer, is related to prognosis, tumor progression, and treatment response. Nevertheless, the correlation of ROS-based molecular signature with clinical outcome and immune cell infiltration has not been thoroughly studied in bladder cancer (BLCA). Accordingly, we aimed to thoroughly examine the role and prognostic value of ROS-related genes in BLCA. Methods We obtained RNA sequencing and clinical data from The Cancer Genome Atlas (TCGA) for bladder cancer (BLCA) patients and identified ROS-associated genes using the GeneCards and Molecular Signatures Database (MSigDB). We then analyzed differential gene expression between BLCA and normal tissues and explored the functions of these ROS-related genes through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Protein-Protein Interaction (PPI) analysis. Prognostic ROS-related genes were identified using Univariate Cox regression (UCR) and LASSO analyses, which were further refined in a Multivariate Cox Regression (MCR) analysis to develop a Prognostic Signature (PS). This PS was validated in the GSE13507 cohort, assessing its predictive power with Kaplan-Meier survival and time-dependent ROC curves. To forecast BLCA outcomes, we constructed a nomogram integrating the PS with clinical variables. We also investigated the signature's molecular characteristics through Gene Set Enrichment Analysis (GSEA), Immune Cell Infiltration (ICI), and Tumor Mutational Burden (TMB) analyses. The Genomics of Drug Sensitivity in Cancer (GDSC) database was used to predict chemotherapy responses based on the PS. Additionally, we screened for Small-Molecule Drugs (SMDs) targeting ROS-related genes using the CMAP database. Finally, we validated our findings by checking protein levels of the signature genes in the Human Protein Atlas (HPA) and confirmed the role of Aldo-keto reductase family 1 member B1 (AKR1B1) through in vitro experiments. Results The constructed and validated PS that comprised 17 ROS-related genes exhibited good performance in predicting overall survival (OS), constituting an independent prognostic biomarker in BLCA patients. Additionally, we successfully established a nomogram with superior predictive capacity, as indicated by the calibration plots. The bioinformatics analysis findings showcased the implication of PS in several oncogenic pathways besides tumor ICI regulation. The PS was negatively associated with the TMB. The high-risk group patients had greater chemotherapy sensitivity in comparison to low-risk group patients. Further, 11 candidate SMDs were identified for treating BLCA. The majority of gene expression exhibited a correlation with the protein expression. In addition, the expression of most genes was consistent with protein expression. Furthermore, to test the gene reliability we constructed, AKR1B1, one of the seventeen genes identified, was used for in-depth validation. In vitro experiments indicate that siRNA-mediated AKR1B1 silencing impeded BLCA cell viability, migration, and proliferation. Conclusions We identified a PS based on 17 ROS-related genes that represented independent OS prognostic factors and 11 candidate SMDs for BLCA treatment, which may contribute to the development of effective individualized therapies for BLCA.
Collapse
Affiliation(s)
- Yulei Li
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Lulu Zhang
- Medical Research Center, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Gang Xu
- Department of Urology, Nanchang People’s Hospital, Nanchang, China
| | - Gang Xu
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Jiajun Chen
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Keyuan Zhao
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Mengyao Li
- Department of Pathology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Jing Jin
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Chao Peng
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Kaifang Wang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, Macao SAR, China
| | - Shouhua Pan
- Department of Urology, Shaoxing People’s Hospital, Zhejiang, Shaoxing, China
| | - Ke Zhu
- Department of Urology, Nanchang People’s Hospital, Nanchang, China
| |
Collapse
|
20
|
Valverde M, Rosales-Cruz P, Torrejon-Gonzalez E, Ponce-Ortiz A, Rodriguez-Sastre MA, Rojas E. Epithelial-Mesenchymal Transition Induced by a Metal Mixture in Liver Cells With Antioxidant Barrier Decreased. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2024; 2024:6983256. [PMID: 39722890 PMCID: PMC11669431 DOI: 10.1155/omcl/6983256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/23/2024] [Indexed: 12/28/2024]
Abstract
Occupational exposure to arsenic (As), cadmium (Cd), and lead (Pb) affects many sectors, necessitating research to understand their transformation mechanisms. In this study, we characterized the process of epithelial-mesenchymal transition (EMT) in a rat hepatic epithelial cell line with decreased expression of catalase and glutamate cysteine ligase catalytic (GCLC) subunit that was exposed to a mixture of As, Cd, and Pb at equimolar occupational exposure concentrations. We evaluated the expression of genes and proteins involved in EMT. Our findings revealed that cells with a decreased antioxidant barrier showed a decreased expression and abundance of epithelial genes when exposed to a mixture of metals. Additionally, we observed alterations in the expression of transcription factors (TFs) associated with EMT and an increase in the expression and abundance of mesenchymal genes. Specifically, we found that E-cadherin expression decreased by ~50% at both the gene and protein levels. In contrast, the expression of vimentin, α-smooth muscle actin, and N-cadherin genes increased by ~70%, whereas their corresponding protein levels increased by nearly 100%. Furthermore, the TFs zinc finger e-box binding homeobox 1 and snail family transcriptional repressor 1 showed a 30% increase in gene expression and an ~80% increase in protein expression. These changes enable the cells to acquire migratory capabilities. Our results confirmed that exposure to this mixture of As, Cd, and Pb can induce EMT in cells with a decreased antioxidant barrier.
Collapse
Affiliation(s)
- M. Valverde
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autonoma de Mexico, Mexico, Mexico
| | - P. Rosales-Cruz
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autonoma de Mexico, Mexico, Mexico
| | - E. Torrejon-Gonzalez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autonoma de Mexico, Mexico, Mexico
| | - A. Ponce-Ortiz
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autonoma de Mexico, Mexico, Mexico
| | - M. A. Rodriguez-Sastre
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autonoma de Mexico, Mexico, Mexico
| | - E. Rojas
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autonoma de Mexico, Mexico, Mexico
| |
Collapse
|
21
|
Stejerean-Todoran I, Gibhardt CS, Bogeski I. Calcium signals as regulators of ferroptosis in cancer. Cell Calcium 2024; 124:102966. [PMID: 39504596 DOI: 10.1016/j.ceca.2024.102966] [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: 10/02/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
The field of ferroptosis research has grown exponentially since this form of cell death was first identified over a decade ago. Ferroptosis, an iron- and ROS-dependent type of cell death, is controlled by various metabolic pathways, including but not limited to redox and calcium (Ca2+) homeostasis, iron fluxes, mitochondrial function and lipid metabolism. Importantly, therapy-resistant tumors are particularly susceptible to ferroptotic cell death, rendering ferroptosis a promising therapeutic strategy against numerous malignancies. Calcium signals are important regulators of both cancer progression and cell death, with recent studies indicating their involvement in ferroptosis. Cells undergoing ferroptosis are characterized by plasma membrane rupture and the formation of nanopores, which facilitate influx of ions such as Ca2+ into the affected cells. Furthermore, mitochondrial Ca²⁺ levels have been implicated in directly influencing the cellular response to ferroptosis. Despite the remarkable progress made in the field, our understanding of the contribution of Ca2+ signals to ferroptosis remains limited. Here, we summarize key connections between Ca²⁺ signaling and ferroptosis in cancer pathobiology and discuss their potential therapeutic significance.
Collapse
Affiliation(s)
- Ioana Stejerean-Todoran
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Christine S Gibhardt
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Ivan Bogeski
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany.
| |
Collapse
|
22
|
Gu Y, Zhao T, Sun B, Zhang Y, Zhang Q, Xu G, Yu C. Integrated gold nanorods-based dual-signal platform for accurate total antioxidant capacity assessment in food samples. Talanta 2024; 280:126650. [PMID: 39128310 DOI: 10.1016/j.talanta.2024.126650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
Accurate assessment of Total Antioxidant Capacity (TAC) in food is crucial for evaluating nutritional quality and potential health benefits. This study aims to enhance the sensitivity and reliability of TAC detection through a dual-signal method, combining colorimetric and photothermal signals. Gold nanorods (AuNRs) were utilized to establish a dual-signal method duo to the colorimetric and photothermal properties. Fenton reaction can etch the AuNRs from the tips, as a result, a blue shift in the longitudinal LSPR absorption peak was obtained, leading to significant changes in color and photothermal effects, facilitating discrimination through both visual observation and thermometer measurements. In the presence of antioxidants, the Fenton reaction was suppressed or inhibited, protecting the AuNRs from etching. The colorimetric and photothermal signals were therefore positively correlated with TAC levels, enabling dual-signal detection of TAC. The linear range of AA was 4-100 μM in both colorimetry and photothermal modes, with detection limits of 1.60 μM and 1.38 μM, respectively. This dual-signal approach achieves low detection limits, enhancing precision and sensitivity. The method thus has the potential to act as a promising candidate for TAC detection in food samples, contributing to improved food quality and safety assessment.
Collapse
Affiliation(s)
- Yuwei Gu
- College of Science, Hebei Agricultural University, Baoding, 071001, PR China
| | - Tengfei Zhao
- Lucky Healthcare Limited Liability Company, Baoding, 071054, PR China
| | - Bo Sun
- College of Mechanical and Electrical Engineering, Hebei Agricultural University, Baoding, 071001, PR China
| | - Yunyi Zhang
- College of Science, Hebei Agricultural University, Baoding, 071001, PR China.
| | - Qingfeng Zhang
- College of Science and Technology, Hebei Agricultural University, Cangzhou, 061100, Hebei, PR China.
| | - Guangcai Xu
- College of Science, Hebei Agricultural University, Baoding, 071001, PR China
| | - Cong Yu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; University of Science and Technology of China, Hefei, 230026, PR China.
| |
Collapse
|
23
|
Chen Z, Yu L, Zheng Z, Wang X, Guo Q, Chen Y, Zhang Y, Zhang Y, Xiao J, Chen K, Fan H, Ding Y. CPT1A mediates radiation sensitivity in colorectal cancer. eLife 2024; 13:RP97827. [PMID: 39607749 PMCID: PMC11604221 DOI: 10.7554/elife.97827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
The prevalence and mortality rates of colorectal cancer (CRC) are increasing worldwide. Radiation resistance hinders radiotherapy, a standard treatment for advanced CRC, leading to local recurrence and metastasis. Elucidating the molecular mechanisms underlying radioresistance in CRC is critical to enhance therapeutic efficacy and patient outcomes. Bioinformatic analysis and tumour tissue examination were conducted to investigate the CPT1A mRNA and protein levels in CRC and their correlation with radiotherapy efficacy. Furthermore, lentiviral overexpression and CRISPR/Cas9 lentiviral vectors, along with in vitro and in vivo radiation experiments, were used to explore the effect of CPT1A on radiosensitivity. Additionally, transcriptomic sequencing, molecular biology experiments, and bioinformatic analyses were employed to elucidate the molecular mechanisms by which CPT1A regulates radiosensitivity. CPT1A was significantly downregulated in CRC and negatively correlated with responsiveness to neoadjuvant radiotherapy. Functional studies suggested that CPT1A mediates radiosensitivity, influencing reactive oxygen species (ROS) scavenging and DNA damage response. Transcriptomic and molecular analyses highlighted the involvement of the peroxisomal pathway. Mechanistic exploration revealed that CPT1A downregulates the FOXM1-SOD1/SOD2/CAT axis, moderating cellular ROS levels after irradiation and enhancing radiosensitivity. CPT1A downregulation contributes to radioresistance in CRC by augmenting the FOXM1-mediated antioxidant response. Thus, CPT1A is a potential biomarker of radiosensitivity and a novel target for overcoming radioresistance, offering a future direction to enhance CRC radiotherapy.
Collapse
Affiliation(s)
- Zhenhui Chen
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhouChina
| | - Lu Yu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Zhihao Zheng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Xusheng Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Qiqing Guo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yuchuan Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Department of Infectious Diseases, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yaowei Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yuqin Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Jianbiao Xiao
- Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical UniversityGuangzhouChina
| | - Keli Chen
- HuiQiao Medical Center, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Hongying Fan
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhouChina
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
24
|
Lee CB, Choi HG, Gurmessa SK, Jang IT, Kumar N, Jiang Z, Kaushik NK, Kim HJ. Enhancing antitumor immunity in Lewis lung cancer through plasma-treated medium-induced activation of dendritic cells. Cancer Cell Int 2024; 24:389. [PMID: 39580412 PMCID: PMC11585098 DOI: 10.1186/s12935-024-03569-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 11/09/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Recently, atmospheric non-thermal plasma jet-treated medium (PTM) has been recognized as a novel strategy in cancer therapy and lymphocyte activation. However, PTM has limitations in inducing a robust antitumor-immune response. This study demonstrated that PTM treatment inhibited tumor progression by activating dendritic cells (DCs). METHOD In this study, we investigated the effects of PTM on selective cytotoxicity and intracellular reactive oxygen species (ROS) generation and oxidative stress-mediated signaling (e.g., glutathione peroxidase, catalase) using respective fluorescence probes in Lewis lung cancer (LLC) cells. Then, the PTM affects the expression of interferon-gamma (IFN)-γ-induced programmed death-ligand 1 (PD-L1) and inhibition of signal transducer and activator of transcription 1 (STAT1) in LLC cells using immunoblotting. Additionally, PTM effects on the tumor cell's death and activation of DCs were done by co-culturing DCs with or without tumor cells. Further, a mouse model was used to evaluate the synergistic antitumor effects of PTM and DCs where tumors are grown under the skin. RESULTS PTM-exposed tumor cells increase intracellular superoxide production, enhancing ROS generation and leading to cancer immunogenic cell death. In addition, PTM suppresses IFN-γ-induced PD-L1 expression and STAT1 activation in tumor cells. The activation of DCs induced by PTM is downregulated when these cells are co-cultured with tumor cells. In vivo, intraperitoneal injection of PTM-activated DCs, as a synergistic agent to intertumoral PTM treatment, led to increased CD4+ and CD8+ T cell infiltration into the tumor and spleen and eventually decreased tumor growth. CONCLUSION Overall, this research introduces a promising avenue for improving lung cancer treatment using PTM to stimulate an immune response and induce cell death in tumor cells. Further studies will be essential to validate these findings and explore clinical applications.
Collapse
Affiliation(s)
- Chae Bok Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, 35015, Korea
| | - Hei Gwon Choi
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, 35015, Korea
- Department of Medical Sciences, Chungnam National University, Daejeon, 35015, Korea
| | - Sintayehu Kebede Gurmessa
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - In-Taek Jang
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, 35015, Korea
| | - Naresh Kumar
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Guwahati (NIPER-G), Kamrup, Assam, 781101, India
| | - Zongyou Jiang
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, 35015, Korea
| | - Nagendra Kumar Kaushik
- Department of Electrical and Biological Physics, Plasma Bioscience Research Center, Kwangwoon University, Seoul, 01897, Korea
| | - Hwa-Jung Kim
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, 35015, Korea.
| |
Collapse
|
25
|
Larrauri-Rodríguez KA, Leon-Chavez BA, Vallejo-Ruiz V, Peña LMP, Maycotte P. Interplay between reactive oxygen species and ERK activation in cervical cancer cells. Front Cell Dev Biol 2024; 12:1465729. [PMID: 39629272 PMCID: PMC11611811 DOI: 10.3389/fcell.2024.1465729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Among the types of cancer affecting women, cervical cancer (CC) is a public health problem with high global incidence and mortality rates. It is currently classified into three main histological types: squamous cell carcinoma (SCC), adenocarcinoma (AC), and adenosquamous (ASC) carcinoma. All of them lack a targeted therapy. The primary risk factor for CC is Human Papilloma Virus (HPV) infection, which is known to increase reactive oxygen species (ROS), contributing to malignant transformation and tumor progression. At basal levels, ROS can function as second messengers in signaling pathways, and elevated concentrations have been linked to their overactivation. One of these, the ERK pathway, is implicated in both cell proliferation and differentiation and is often dysregulated in cancer, promoting malignant transformation. Several studies have proposed antioxidant supplementation or ERK inhibitors as potential therapies. Methods In vitro studies were performed using CC cell lines. ROS levels were evaluated by flow cytometry; cellular proliferation, death and migration were evaluated using real-time microscopy; cell viability was evaluated with crystal violet staining, and phosphorylated ERK levels were evaluated by Western Blot. A bioinformatic analysis was done in a cervical cancer database. Results We elucidate part of the complex interplay between ROS and ERK pathway in CC pro-tumorigenic characteristics. Through bioinformatic analysis, we found distinct ROS and ERK activation patterns across CC tumor samples from different histological types. However, in vitro, ROS regulated migration and viability in CC, with no discernible variance based on histological classification. ERK activation, however, differed according to the histological type with SCC displaying increased ERK activation compared to AC and regulating cellular migration in SCC cells. Discussion Our study identifies a potential synergistic interaction between ROS and ERK inhibitors, highlighting the therapeutic promise of combinatorial targeting for CC treatment. These findings underscore the importance of personalized approaches aimed at improving the outcomes of CC patients.
Collapse
Affiliation(s)
- Karen Andrea Larrauri-Rodríguez
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), OOAD Puebla, Puebla, Mexico
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla (BUAP), Ciudad Universitaria, Puebla, Mexico
| | - Bertha Alicia Leon-Chavez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla (BUAP), Ciudad Universitaria, Puebla, Mexico
| | - Verónica Vallejo-Ruiz
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), OOAD Puebla, Puebla, Mexico
| | - Lourdes Millán-Perez Peña
- Centro de Química, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Ciudad Universitaria, Puebla, Mexico
| | - Paola Maycotte
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), OOAD Puebla, Puebla, Mexico
| |
Collapse
|
26
|
Mohammadi Zonouz A, Taghavi S, Nekooei S, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. Synthesis of targeted doxorubicin-loaded gold nanorod -mesoporous manganese dioxide core-shell nanostructure for ferroptosis, chemo-photothermal therapy in vitro and in vivo. Int J Pharm 2024; 665:124725. [PMID: 39293581 DOI: 10.1016/j.ijpharm.2024.124725] [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: 08/01/2024] [Revised: 09/14/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
In the current study, a core-shell inorganic nanostructure comprising a gold nanorod core and -mesoporous manganese dioxide shell was synthesized. Then, the mesoporous manganese dioxide shell was loaded with doxorubicin (DOX) and then coated with pluronic F127 and pluronic F127-folic acid conjugate (1.5:1 wt ratio of pluronic F127: pluronic F127-folic acid conjugate) to prepare targeted final platform. In this design, mesoporous manganese dioxide acted as a reservoir for DOX loading, anti-hypoxia, and MRI contrast agent, while the gold nanorod core acted as a photothermal and CT scan imaging agent. DOX was encapsulated in the mesoporous manganese dioxide shell with a loading capacity and loading efficiency of 19.8 % ± 0.2 and 99.0 % ± 0.9, respectively. The in vitro release experiment showed the impact of glutathione (GSH), mildly acidic pH, and laser irradiating toward accelerated stimuli-responsive DOX release. The ·OH production of the prepared platform was verified by methylene blue (MB) decomposition reaction. Furthermore, thermal imaging exhibited the ability of the prepared platform to convert the NIR irradiation to heat. In vitro cytotoxicity tests on the folate receptor-positive 4 T1 cell line revealed the remarkable cytotoxicity of the targeted formulation compared to the nontargeted formulation (statistically significant). The MTT experiment demonstrated that exposure to laser 808 irradiation enhanced cytotoxicity of the targeted formulation (p < 0.0001). The production of ROS in 4 T1 cells following treatment with the targeted formulation was demonstrated by the dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay. Furthermore, in vivo investigations by implementing subcutaneous 4 T1 tumorized female BABL/c mice indicated that the prepared platform was an effective system in suppressing tumor growth by combining chemotherapy with PTT (photothermal therapy). Additionally, simultanous PTT and anti-hypoxic activity of this system showed potent tumor growth suppression impact. The percent of tumor size reduction in mice treated with FA-F127-DOX@Au-MnO2 + 808 nm laser compared to the control group was 99.7 %. The results of the biodistribution investigation showed tumor accumulation and modified pharmacokinetics of the targeted system. Lastly, 6 and 24 h post-intravenous injection, CT-scan and MR imagings capability of the prepared platform was verified in preclinical stage. The prepared multipurpose system introduces great opportunity to provide multiple treatment strategy along with multimodal imaging capability in a single platform for breast cancer treatment.
Collapse
Affiliation(s)
- Aidin Mohammadi Zonouz
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Taghavi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medicinal Chemistry Department, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Zhang Z, Chen C, Li X, Zheng J, Zhao Y. Regulation of leukemogenesis via redox metabolism. Trends Cell Biol 2024; 34:928-941. [PMID: 39492031 DOI: 10.1016/j.tcb.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 11/05/2024]
Abstract
Redox metabolism plays a central role in the cellular metabolism network, involves catabolic and anabolic reactions of diverse biomass, and determines the redox state of cells. It can be quantitatively and conveniently measured in living cells and organisms with genetically encoded fluorescent sensors, providing novel insights that cannot be readily acquired via conventional metabolic assays. Here, we review the recent progress on the regulation of leukemogenesis via redox metabolism, especially redox biosensor-based findings. In general, low reactive oxygen species levels and high reductive capacity promote leukemogenesis and chemotherapy resistance in leukemia cells, and acute leukemia cells rewire metabolism of glucose, fatty acids, and some amino acids, together with oxidative phosphorylation, to fuel energy production, support biomass-related synthesis, and survive oxidative stress. In summary, redox metabolism is a potential target for the development of novel therapies for leukemia or beneficial dietary regimens for patients with refractory and relapsed leukemia.
Collapse
Affiliation(s)
- Zhuo Zhang
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Chiqi Chen
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xie Li
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yuzheng Zhao
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
28
|
Xiao W, Xu C. Cystine/cysteine metabolism regulates the progression and response to treatment of triple‑negative breast cancer (Review). Oncol Lett 2024; 28:521. [PMID: 39268159 PMCID: PMC11391256 DOI: 10.3892/ol.2024.14654] [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: 02/08/2024] [Accepted: 07/04/2024] [Indexed: 09/15/2024] Open
Abstract
Breast cancer is the most prevalent neoplasm affecting women globally, of which a notable proportion of cases are triple-negative breast cancer (TNBC). However, there are limited curative treatment options for patients with TNBC, despite advancements in the field. Amino acids and amino acid transporters serve vital roles in the regulation of tumor metabolism. Notably, cystine and cysteine can interconvert via a redox reaction, with cysteine exerting control on cell survival and growth and exogenous cystine serving a crucial role in the proliferation of numerous types of cancers. Breast cancer has been reported to disrupt the cystine/cysteine metabolism pathway, as cystine and cysteine transporters affect the development and growth of tumors. The present review aims to provide a comprehensive overview of the metabolic pathways involving cystine and cysteine in normal and TNBC cells. Furthermore, the roles of cystine and cysteine transporters in TNBC progression and metastasis and their potential as therapeutic targets for treatment of TNBC are evaluated.
Collapse
Affiliation(s)
- Wanting Xiao
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Chaoyang Xu
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| |
Collapse
|
29
|
Liao W, Cheng B, Wang C, Richardson JJ, Naito M, Miyata K, Ejima H. Surface-Initiated Synergistic Disassembly of Metal-Phenolic Networks by Redox and Hydrolytic Reactions. CHEMISTRY OF MATERIALS 2024; 36:9646-9657. [DOI: 10.1021/acs.chemmater.4c01724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Affiliation(s)
- Wenting Liao
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Bohan Cheng
- Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM2), Hiroshima University, Hiroshima 739-0046, Japan
- RIKEN Center of Emergent Matter Science (CEMS), Saitama 739-0046, Japan
| | - Chenyu Wang
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Joseph J. Richardson
- Department of Chemical and Environmental Engineering, RMIT University, Melbourne, Victoria 3000, Australia
| | - Mitsuru Naito
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kanjiro Miyata
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Hirotaka Ejima
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
30
|
Yang J, Fu Q, Jiang H, Zhong H, Qin HK, Miao X, Li Y, Liu M, Yao J. Blue light photobiomodulation induced osteosarcoma cell death by facilitating ferroptosis and eliciting an incomplete tumor cell stress response. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 258:113003. [PMID: 39121719 DOI: 10.1016/j.jphotobiol.2024.113003] [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: 05/09/2024] [Revised: 07/10/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
To investigate the potential of blue light photobiomodulation (PBM) in inducing ferroptosis, a novel form of regulated cell death, in OS cells, considering its known effectiveness in various cancer models. In this investigation, we exposed human OS cell lines, HOS and MG63, to different wavelengths (420, 460 and 480 nm) of blue light at varying irradiances, and examined cellular responses such as viability, apoptosis, levels of reactive oxygen species (ROS), and mitochondrial membrane potential (MMP). Transcriptome sequencing was employed to unravel the molecular mechanisms underlying blue light-induced effects, with validation via quantitative real-time PCR (qRT-PCR). Our findings revealed a wavelength- and time-dependent decrease in cell viability, accompanied by increased apoptosis and oxidative stress. Transcriptomic analysis identified differential expression of genes associated with ferroptosis, oxidative stress, and iron metabolism, further validated by qRT-PCR. These results implicated ferroptosis as a significant mechanism in the blue light-induced death of OS cells, potentially mediated by ROS generation and disruption of iron homeostasis. Also, An incomplete stress response was observed in MG63 cells induced by blue light exposure. Hence, blue light PBM holds promise as a therapeutic approach in OS clinical investigations; however, additional exploration of its underlying mechanisms remains imperative.
Collapse
Affiliation(s)
- Jiali Yang
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China
| | - Qiqi Fu
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China
| | - Hui Jiang
- Academy for Engineering and Technology, Fudan University, 220th Handan Road, Shanghai 200433, China
| | - Hongyu Zhong
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third School of Clinical Medicine, The Third Affiliated Hospital of Southern Medical University, No.183, Zhongshan Avenue West, Guangzhou 510515, China
| | - Hao Kuan Qin
- Academy for Engineering and Technology, Fudan University, 220th Handan Road, Shanghai 200433, China
| | - Xiaojing Miao
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China
| | - Yinghua Li
- Shanghai Fifth People's Hospital, Fudan University, 801th Heqing Road, Shanghai 200240, China.
| | - Muqing Liu
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China; Zhongshan Fudan Joint Innovation Center, 6th Xiangxing Road, Zhongshan 28403, China.
| | - Jinghui Yao
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third School of Clinical Medicine, The Third Affiliated Hospital of Southern Medical University, No.183, Zhongshan Avenue West, Guangzhou 510515, China.
| |
Collapse
|
31
|
Zhang Y, Lei F, Qian W, Zhang C, Wang Q, Liu C, Ji H, Liu Z, Wang F. Designing intelligent bioorthogonal nanozymes: Recent advances of stimuli-responsive catalytic systems for biomedical applications. J Control Release 2024; 373:929-951. [PMID: 39097195 DOI: 10.1016/j.jconrel.2024.07.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Bioorthogonal nanozymes have emerged as a potent tool in biomedicine due to their unique ability to perform enzymatic reactions that do not interfere with native biochemical processes. The integration of stimuli-responsive mechanisms into these nanozymes has further expanded their potential, allowing for controlled activation and targeted delivery. As such, intelligent bioorthogonal nanozymes have received more and more attention in developing therapeutic approaches. This review provides a comprehensive overview of the recent advances in the development and application of stimuli-responsive bioorthogonal nanozymes. By summarizing the design outlines for anchoring bioorthogonal nanozymes with stimuli-responsive capability, this review seeks to offer valuable insights and guidance for the rational design of these remarkable materials. This review highlights the significant progress made in this exciting field with different types of stimuli and the various applications. Additionally, it also examines the current challenges and limitations in the design, synthesis, and application of these systems, and proposes potential solutions and research directions. This review aims to stimulate further research toward the development of more efficient and versatile stimuli-responsive bioorthogonal nanozymes for biomedical applications.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Fang Lei
- School of Public Health, Nantong University, Nantong 226019, China
| | - Wanlong Qian
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Chengfeng Zhang
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Qi Wang
- School of Public Health, Nantong University, Nantong 226019, China
| | - Chaoqun Liu
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Haiwei Ji
- School of Public Health, Nantong University, Nantong 226019, China
| | - Zhengwei Liu
- Precision Immunology Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York 10029, USA.
| | - Faming Wang
- School of Public Health, Nantong University, Nantong 226019, China.
| |
Collapse
|
32
|
Kump DS. Mechanisms Underlying the Rarity of Skeletal Muscle Cancers. Int J Mol Sci 2024; 25:6480. [PMID: 38928185 PMCID: PMC11204341 DOI: 10.3390/ijms25126480] [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: 05/07/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Skeletal muscle (SKM), despite comprising ~40% of body mass, rarely manifests cancer. This review explores the mechanisms that help to explain this rarity, including unique SKM architecture and function, which prohibits the development of new cancer as well as negates potential metastasis to SKM. SKM also presents a unique immune environment that may magnify the anti-tumorigenic effect. Moreover, the SKM microenvironment manifests characteristics such as decreased extracellular matrix stiffness and altered lactic acid, pH, and oxygen levels that may interfere with tumor development. SKM also secretes anti-tumorigenic myokines and other molecules. Collectively, these mechanisms help account for the rarity of SKM cancer.
Collapse
Affiliation(s)
- David S Kump
- Department of Biological Sciences, Winston-Salem State University, 601 Martin Luther King Jr. Dr., Winston-Salem, NC 27110, USA
| |
Collapse
|
33
|
Fnu G, Weber GF. Targeting the core program of metastasis with a novel drug combination. Cancer Med 2024; 13:e7291. [PMID: 38826119 PMCID: PMC11145026 DOI: 10.1002/cam4.7291] [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: 10/10/2023] [Revised: 04/29/2024] [Accepted: 05/04/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND We previously reported that metastases are generally characterized by a core program of gene expression that activates tissue remodeling/vascularization, alters ion homeostasis, induces the oxidative metabolism, and silences extracellular matrix interactions. This core program distinguishes metastases from their originating primary tumors as well as from their destination host tissues. Therefore, the gene products involved are potential targets for anti-metastasis drug treatment. METHODS Because the silencing of extracellular matrix interactions predisposes to anoiks in the absence of active survival mechanisms, we tested inhibitors against the other three components. RESULTS Individually, the low-specificity VEGFR blocker pazopanib (in vivo combined with marimastat), the antioxidant dimethyl sulfoxide (or the substitute atovaquone, which is approved for internal administration), and the ionic modulators bumetanide and tetrathiomolybdate inhibited soft agar colony formation by breast and pancreatic cancer cell lines. The individual candidate agents have a record of use in humans (with limited efficacy when administered individually) and are available for repurposing. In combination, the effects of these drugs were additive or synergistic. In two mouse models of cancer (utilizing 4T1 cells or B16-F10 cells), the combination treatment with these medications, applied immediately (to prevent metastasis formation) or after a delay (to suppress established metastases), dramatically reduced the occurrence of disseminated foci. CONCLUSIONS The combination of tissue remodeling inhibitors, suppressors of the oxidative metabolism, and ion homeostasis modulators has very strong promise for the treatment of metastases by multiple cancers.
Collapse
Affiliation(s)
- Gulimirerouzi Fnu
- James L. Winkle College of PharmacyUniversity of Cincinnati Academic Health CenterCincinnatiOhioUSA
| | - Georg F. Weber
- James L. Winkle College of PharmacyUniversity of Cincinnati Academic Health CenterCincinnatiOhioUSA
| |
Collapse
|
34
|
Tripathy S, Haque S, Londhe S, Das S, Norbert CC, Chandra Y, Sreedhar B, Patra CR. ROS mediated Cu[Fe(CN) 5NO] nanoparticles for triple negative breast cancer: A detailed study in preclinical mouse model. BIOMATERIALS ADVANCES 2024; 160:213832. [PMID: 38547763 DOI: 10.1016/j.bioadv.2024.213832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 05/04/2024]
Abstract
Triple negative breast cancer (TNBC) is an aggressive form of tumor, more prevalent in younger women resulting in poor survival rate (2nd in cancer deaths) because of its asymptomatic existence. The most popular and convenient approach for the treatment of TNBC is chemotherapy which is associated with several limitations. Considering the importance of nanotechnology in health care system, in the present manuscript, we have designed and developed a simple, efficient, cost effective, and ecofriendly method for the synthesis of copper nitroprusside analogue nanoparticles (Cu[Fe(CN)5NO] which is abbreviated as CuNPANP that may be the potential anti-cancer nanomedicine for the treatment of TNBC. Copper (present in CuNPANP) is used because of its affordability, nutritional value and various biomedical applications. The CuNPANP are thoroughly characterized using several analytical techniques. The in vitro cell viability (in normal cells) and the ex vivo hemolysis assay reveal the biocompatible nature of CuNPANP. The anti-cancer activity of the CuNPANP is established in TNBC cells (MDA-MB-231 and 4T1) through several in vitro assays along with plausible mechanisms. The intraperitoneal administration of CuNPANP in orthotopic breast tumor model by transplanting 4T1 cells into the mammary fat pad of BALB/c mouse significantly inhibits the growth of breast carcinoma as well as increases the survival time of tumor-bearing mice. These results altogether potentiate the anti-cancer efficacy of CuNPANP as a smart therapeutic nanomedicine for treating TNBC in near future after bio-safety evaluation in large animals.
Collapse
Affiliation(s)
- Sanchita Tripathy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Shagufta Haque
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Swapnali Londhe
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Sourav Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Caroline Celine Norbert
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India
| | - Yogesh Chandra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Bojja Sreedhar
- Department of Analytical & Structural ChemistryCSIR-Indian Institute of Chemical Technology, Uppal Road,Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India.
| |
Collapse
|
35
|
Liu X, Bai Y, Zhou B, Yao W, Song S, Liu J, Zheng C. Recent advances in hepatocellular carcinoma-targeted nanoparticles. Biomed Mater 2024; 19:042004. [PMID: 38697209 DOI: 10.1088/1748-605x/ad46d3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/01/2024] [Indexed: 05/04/2024]
Abstract
In the field of medicine, we often brave the unknown like interstellar explorers, especially when confronting the formidable opponent of hepatocellular carcinoma (HCC). The global burden of HCC remains significant, with suboptimal treatment outcomes necessitating the urgent development of novel drugs and treatments. While various treatments for liver cancer, such as immunotherapy and targeted therapy, have emerged in recent years, improving their transport and therapeutic efficiency, controlling their targeting and release, and mitigating their adverse effects remains challenging. However, just as we grope through the darkness, a glimmer of light emerges-nanotechnology. Recently, nanotechnology has attracted attention because it can increase the local drug concentration in tumors, reduce systemic toxicity, and has the potential to enhance the effectiveness of precision therapy for HCC. However, there are also some challenges hindering the clinical translation of drug-loaded nanoparticles (NPs). Just as interstellar explorers must overcome interstellar dust, we too must overcome various obstacles. In future researches, the design and development of nanodelivery systems for novel drugs treating HCC should be the first attention. Moreover, researchers should focus on the active targeting design of various NPs. The combination of the interventional therapies and drug-loaded NPs will greatly advance the process of precision HCC therapy.
Collapse
Affiliation(s)
- Xiaoming Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Yaowei Bai
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Binqian Zhou
- Department of Ultrasound, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, People's Republic of China
| | - Wei Yao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Songlin Song
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Jiacheng Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| |
Collapse
|
36
|
Li X, Gao J, Wu C, Wang C, Zhang R, He J, Xia ZJ, Joshi N, Karp JM, Kuai R. Precise modulation and use of reactive oxygen species for immunotherapy. SCIENCE ADVANCES 2024; 10:eadl0479. [PMID: 38748805 PMCID: PMC11095489 DOI: 10.1126/sciadv.adl0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/10/2024] [Indexed: 05/19/2024]
Abstract
Reactive oxygen species (ROS) play an important role in regulating the immune system by affecting pathogens, cancer cells, and immune cells. Recent advances in biomaterials have leveraged this mechanism to precisely modulate ROS levels in target tissues for improving the effectiveness of immunotherapies in infectious diseases, cancer, and autoimmune diseases. Moreover, ROS-responsive biomaterials can trigger the release of immunotherapeutics and provide tunable release kinetics, which can further boost their efficacy. This review will discuss the latest biomaterial-based approaches for both precise modulation of ROS levels and using ROS as a stimulus to control the release kinetics of immunotherapeutics. Finally, we will discuss the existing challenges and potential solutions for clinical translation of ROS-modulating and ROS-responsive approaches for immunotherapy, and provide an outlook for future research.
Collapse
Affiliation(s)
- Xinyan Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jingjing Gao
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Biomedical Engineering, Material Science and Engineering Graduate Program and The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Chengcheng Wu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Chaoyu Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ruoshi Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jia He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ziting Judy Xia
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nitin Joshi
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey M. Karp
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
37
|
Cai H, Meng Z, Yu F. The involvement of ROS-regulated programmed cell death in hepatocellular carcinoma. Crit Rev Oncol Hematol 2024; 197:104361. [PMID: 38626849 DOI: 10.1016/j.critrevonc.2024.104361] [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: 11/04/2023] [Revised: 03/11/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024] Open
Abstract
Reactive oxidative species (ROS) is a crucial factor in the regulation of cellular biological activity and function, and aberrant levels of ROS can contribute to the development of a variety of diseases, particularly cancer. Numerous discoveries have affirmed that this process is strongly associated with "programmed cell death (PCD)," which refers to the suicide protection mechanism initiated by cells in response to external stimuli, such as apoptosis, autophagy, ferroptosis, etc. Research has demonstrated that ROS-induced PCD is crucial for the development of hepatocellular carcinoma (HCC). These activities serve a dual function in both facilitating and inhibiting cancer, suggesting the existence of a delicate balance within healthy cells that can be disrupted by the abnormal generation of reactive oxygen species (ROS), thereby influencing the eventual advancement or regression of a tumor. In this review, we summarize how ROS regulates PCD to influence the tumorigenesis and progression of HCC. Studying how ROS-induced PCD affects the progression of HCC at a molecular level can help develop better prevention and treatment methods and facilitate the design of more effective preventative and therapeutic strategies.
Collapse
Affiliation(s)
- Hanchen Cai
- The First Afliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Ziqi Meng
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China; The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Fujun Yu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
38
|
Zhao NN, Wang Q, Yang DM, Li DL, Han Y, Zhao S, Zou X, Zhang CY. Elongation and Ligation-Mediated Differential Coding for Label-Free and Locus-Specific Analysis of 8-Oxo-7,8-dihydroguanine in DNA. Anal Chem 2024; 96:5323-5330. [PMID: 38501982 DOI: 10.1021/acs.analchem.4c00387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Oxidative DNA damage is closely associated with the occurrence of numerous human diseases and cancers. 8-Oxo-7,8-dihydroguanine (8-oxoG) is the most prevalent form of DNA damage, and it has become not only an oxidative stress biomarker but also a new epigenetic-like biomarker. However, few approaches are available for the locus-specific detection of 8-oxoG because of the low abundance of 8-oxoG damage in DNA and the limited sensitivity of existing assays. Herein, we demonstrate the elongation and ligation-mediated differential coding for label-free and locus-specific analysis of 8-oxoG in DNA. This assay is very simple without the involvement of any specific labeled probes, complicated steps, and large sample consumption. The utilization of Bsu DNA polymerase can specifically initiate a single-base extension reaction to incorporate dATP into the opposite position of 8-oxoG, endowing this assay with excellent selectivity. The introduction of cascade amplification reaction significantly enhances the sensitivity. The proposed method can monitor 8-oxoG with a limit of detection of 8.21 × 10-19 M (0.82 aM), and it can identify as low as 0.001% 8-oxoG damage from a complex mixture with excessive undamaged DNAs. This method can be further applied to measure 8-oxoG levels in the genomic DNA of human cells under diverse oxidative stress, holding prospect potential in the dynamic monitoring of critical 8-oxoG sites, early clinical diagnosis, and gene damage-related biomedical research.
Collapse
Affiliation(s)
- Ning-Ning Zhao
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Qian Wang
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Dong-Ming Yang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, Ministry of Education, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Dong-Ling Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yun Han
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Shulin Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, Ministry of Education, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Xiaoran Zou
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Chun-Yang Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
39
|
Cavanagh RJ, Monteiro PF, Moloney C, Travanut A, Mehradnia F, Taresco V, Rahman R, Martin SG, Grabowska AM, Ashford MB, Alexander C. Free drug and ROS-responsive nanoparticle delivery of synergistic doxorubicin and olaparib combinations to triple negative breast cancer models. Biomater Sci 2024; 12:1822-1840. [PMID: 38407276 DOI: 10.1039/d3bm01931d] [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: 02/27/2024]
Abstract
Combinations of the topoisomerase II inhibitor doxorubicin and the poly (ADP-ribose) polymerase inhibitor olaparib offer potential drug-drug synergy for the treatment of triple negative breast cancers (TNBC). In this study we performed in vitro screening of combinations of these drugs, administered directly or encapsulated within polymer nanoparticles, in both 2D and in 3D spheroid models of breast cancer. A variety of assays were used to evaluate drug potency, and calculations of combination index (CI) values indicated that synergistic effects of drug combinations occurred in a molar-ratio dependent manner. It is suggested that the mechanisms of synergy were related to enhancement of DNA damage as shown by the level of double-strand DNA breaks, and mechanisms of antagonism associated with mitochondrial mediated cell survival, as indicated by reactive oxygen species (ROS) generation. Enhanced drug delivery and potency was observed with nanoparticle formulations, with a greater extent of doxorubicin localised to cell nuclei as evidenced by microscopy, and higher cytotoxicity at the same time points compared to free drugs. Together, the work presented identifies specific combinations of doxorubicin and olaparib which were most effective in a panel of TNBC cell lines, explores the mechanisms by which these combined agents might act, and shows that formulation of these drug combinations into polymeric nanoparticles at specific ratios conserves synergistic action and enhanced potency in vitro compared to the free drugs.
Collapse
Affiliation(s)
| | - Patrícia F Monteiro
- School of Pharmacy, University of Nottingham, NG7 2RD, UK.
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - Cara Moloney
- School of Pharmacy, University of Nottingham, NG7 2RD, UK.
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | | | | | | | - Ruman Rahman
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Stewart G Martin
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Anna M Grabowska
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Marianne B Ashford
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | | |
Collapse
|
40
|
Zhang F, Xiang Y, Ma Q, Guo E, Zeng X. A deep insight into ferroptosis in lung disease: facts and perspectives. Front Oncol 2024; 14:1354859. [PMID: 38562175 PMCID: PMC10982415 DOI: 10.3389/fonc.2024.1354859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
In the last decade, ferroptosis has received much attention from the scientific research community. It differs from other modes of cell death at the morphological, biochemical, and genetic levels. Ferroptosis is mainly characterized by non-apoptotic iron-dependent cell death caused by iron-dependent lipid peroxide excess and is accompanied by abnormal iron metabolism and oxidative stress. In recent years, more and more studies have shown that ferroptosis is closely related to the occurrence and development of lung diseases. COPD, asthma, lung injury, lung fibrosis, lung cancer, lung infection and other respiratory diseases have become the third most common chronic diseases worldwide, bringing serious economic and psychological burden to people around the world. However, the exact mechanism by which ferroptosis is involved in the development and progression of lung diseases has not been fully revealed. In this manuscript, we describe the mechanism of ferroptosis, targeting of ferroptosis related signaling pathways and proteins, summarize the relationship between ferroptosis and respiratory diseases, and explore the intervention and targeted therapy of ferroptosis for respiratory diseases.
Collapse
Affiliation(s)
- Fan Zhang
- Wuhan University of Science and Technology, School of Medicine, Wuhan, China
| | - Yu Xiang
- Wuhan University of Science and Technology, School of Medicine, Wuhan, China
| | - Qiao Ma
- Wuhan University of Science and Technology, School of Medicine, Wuhan, China
| | - E. Guo
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Xiansheng Zeng
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
41
|
Zhu C, Lu Y, Wang S, Song J, Ding Y, Wang Y, Dong C, Liu J, Qiu W, Qi W. Nortriptyline hydrochloride, a potential candidate for drug repurposing, inhibits gastric cancer by inducing oxidative stress by triggering the Keap1-Nrf2 pathway. Sci Rep 2024; 14:6050. [PMID: 38480798 PMCID: PMC10937941 DOI: 10.1038/s41598-024-56431-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/06/2024] [Indexed: 03/17/2024] Open
Abstract
Effective drugs for the treatment of gastric cancer (GC) are still lacking. Nortriptyline Hydrochloride (NTP), a commonly used antidepressant medication, has been demonstrated by numerous studies to have antitumor effects. This study first validated the ability of NTP to inhibit GC and preliminarily explored its underlying mechanism. To begin with, NTP inhibits the activity of AGS and HGC27 cells (Human-derived GC cells) in a dose-dependent manner, as well as proliferation, cell cycle, and migration. Moreover, NTP induces cell apoptosis by upregulating BAX, BAD, and c-PARP and downregulating PARP and Bcl-2 expression. Furthermore, the mechanism of cell death caused by NTP is closely related to oxidative stress. NTP increases intracellular reactive oxygen species (ROS) and malondialdehyde (MDA) levels, decreasing the mitochondrial membrane potential (MMP) and inducing glucose (GSH) consumption. While the death of GC cells can be partially rescued by ROS inhibitor N-acetylcysteine (NAC). Mechanistically, NTP activates the Kelch-like ECH-associated protein (Keap1)-NF-E2-related factor 2 (Nrf2) pathway, which is an important pathway involved in oxidative stress. RNA sequencing and proteomics analysis further revealed molecular changes at the mRNA and protein levels and provided potential targets and pathways through differential gene expression analysis. In addition, NTP can inhibited tumor growth in nude mouse subcutaneous tumor models constructed respectively using AGS and MFC (mouse-derived GC cells), providing preliminary evidence of its effectiveness in vivo. In conclusion, our study demonstrated that NTP exhibits significant anti-GC activity and is anticipated to be a candidate for drug repurposing.
Collapse
Affiliation(s)
- Chunyang Zhu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yangyang Lu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shasha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jialin Song
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yixin Ding
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chen Dong
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jiani Liu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wensheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Weiwei Qi
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
42
|
Brahma D, Sarangi AN, Kaushik R, Gupta AN. Oxidative stress induced conformational changes of human serum albumin. Phys Chem Chem Phys 2024; 26:8528-8538. [PMID: 38411624 DOI: 10.1039/d4cp00059e] [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: 02/28/2024]
Abstract
Oxidative stress, generated by reactive oxygen species (ROS), is responsible for the loss of structure and functionality of proteins and is associated with several aging-related diseases. Here, we report an in vitro study to gauge the effect of ROS on the structural rearrangement of human serum albumin (HSA), a plasma protein, through metal-catalyzed oxidation (MCO) at physiological temperature through various biophysical techniques like UV-vis absorption, circular dichroism (CD), differential scanning calorimetry (DSC), MALDI-TOF, FTIR, and Raman spectroscopy. The UV-vis spectra of oxidized HSA show an early blueshift, signifying the unfolding of the protein because of ROS followed by the broadening of the absorption peak at a longer time. The DSC data corroborate the observation, revealing an exothermic transition for the oxidized sample at a longer time, suggesting in situ aggregation. The CD and FTIR spectra indicate the associated secondary structural changes occurring with time, depicting the variation of the helical content of HSA. The amide-III analysis of Raman data also complements the structural changes, and MALDI-TOF data show the mass distribution with time. Overall, this work might help determine the effect of oxidation on the biological activity of serum albumin as it can impact the physiological properties of HSA.
Collapse
Affiliation(s)
- Debdip Brahma
- Biophysics and Soft Matter Laboratory, Department of Physics, Indian Institute of Technology, Kharagpur, 721302, India.
| | - Akshay Narayan Sarangi
- Biophysics and Soft Matter Laboratory, Department of Physics, Indian Institute of Technology, Kharagpur, 721302, India.
| | - Rupal Kaushik
- Biophysics and Soft Matter Laboratory, Department of Physics, Indian Institute of Technology, Kharagpur, 721302, India.
| | - Amar Nath Gupta
- Biophysics and Soft Matter Laboratory, Department of Physics, Indian Institute of Technology, Kharagpur, 721302, India.
| |
Collapse
|
43
|
Cao Y, Hou L, Li M, Zhang J, Wang L, Liu C, Luo T, Yan L, Zheng L. Broccoli extracellular vesicles enhance the therapeutic effects and restore the chemosensitivity of 5-fluorouracil on colon cancer. Food Chem Toxicol 2024; 186:114563. [PMID: 38442787 DOI: 10.1016/j.fct.2024.114563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/19/2024] [Accepted: 02/29/2024] [Indexed: 03/07/2024]
Abstract
Broccoli contains an amount of biologically active substances, which bring beneficial effects on human health. Plant extracellular vesicles have been shown to be novel key factors in cancer diagnosis and tumor therapy. To date, the challenge of overcoming chemoresistance to 5-fluorouracil (5-FU) to facilitate the clinical management of colorectal cancer (CRC) has not been successful. Nevertheless, the functions of broccoli extracellular vesicles (BEVs) in the progression of CRC and 5-FU resistance are predominantly unclear. Herein, we showed that BEVs isolated from broccoli juice were effectively taken up by colorectal cancer HT-29 cells. The co-administration of BEVs and 5-FU significantly inhibited the proliferation and migration of colorectal cancer HT-29 cells, effectively blocking cell cycle progression. Furthermore, the co-administration of BEVs and 5-FU induced apoptosis by stimulating ROS production and disrupting mitochondrial function. Importantly, we found that BEVs reversed 5-FU resistance in HT-29 cells by suppressing the abnormal activation of the PI3K/Akt/mTOR signaling pathway. Collectively, our findings represent a novel strategy for utilizing BEVs to improve the efficacy of colorectal cancer treatment and enhance 5-FU chemosensitivity.
Collapse
Affiliation(s)
- Yaqi Cao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Linhai Hou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Meiqi Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Jing Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Lei Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Changhong Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Tianyu Luo
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Ling Yan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China; Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei, 230009, China.
| | - Lei Zheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China; Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei, 230009, China; Research Laboratory of Agricultural Environment and Food Safety, Anhui Modern Agricultural Industry Technology System, Hefei, 230009, China.
| |
Collapse
|
44
|
Najeeb HA, Sanusi T, Saldanha G, Brown K, Cooke MS, Jones GD. Redox modulation of oxidatively-induced DNA damage by ascorbate enhances both in vitro and ex-vivo DNA damage formation and cell death in melanoma cells. Free Radic Biol Med 2024; 213:309-321. [PMID: 38262545 DOI: 10.1016/j.freeradbiomed.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/14/2024] [Indexed: 01/25/2024]
Abstract
Elevated genomic instability in cancer cells suggests a possible model-scenario for their selective killing via the therapeutic delivery of well-defined levels of further DNA damage. To examine this scenario, this study investigated the potential for redox modulation of oxidatively-induced DNA damage by ascorbate in malignant melanoma (MM) cancer cells, to selectively enhance both DNA damage and MM cell killing. DNA damage was assessed by Comet and ɣH2AX assays, intracellular oxidising species by dichlorofluorescein fluorescence, a key antioxidant enzymatic defence by assessment of catalase activity and cell survival was determined by clonogenic assay. Comet revealed that MM cells had higher endogenous DNA damage levels than normal keratinocytes (HaCaT cells); this correlated MM cells having higher intracellular oxidising species and lower catalase activity, and ranked with MM cell melanin pigmentation. Comet also showed MM cells more sensitive towards the DNA damaging effects of exogenous H2O2, and that ascorbate further enhanced this H2O2-induced damage in MM cells; again, with MM cell sensitivity to induced damage ranking with degree of cell pigmentation. Furthermore, cell survival data indicated that ascorbate enhanced H2O2-induced clonogenic cell death selectively in MM cells whilst protecting HaCaT cells. Finally, we show that ascorbate serves to enhance the oxidising effects of the MM therapeutic drug Elesclomol in both established MM cells in vitro and primary cell cultures ex vivo. Together, these results suggest that ascorbate selectively enhances DNA damage and cell-killing in MM cells. This raises the option of incorporating ascorbate into clinical oxidative therapies to treat MM.
Collapse
Affiliation(s)
- Hishyar A Najeeb
- Leicester Cancer Research Centre, Department of Genetics & Genome Biology, University of Leicester, UK
| | - Timi Sanusi
- Leicester Medical School, University of Leicester, UK
| | - Gerald Saldanha
- University Hospitals of Leicester NHS Trust, Leicester Royal Infirmary, UK
| | - Karen Brown
- Leicester Cancer Research Centre, Department of Genetics & Genome Biology, University of Leicester, UK
| | - Marcus S Cooke
- Oxidative Stress Group, Department of Molecular Biosciences, University of South Florida, USA.
| | - George Dd Jones
- Leicester Cancer Research Centre, Department of Genetics & Genome Biology, University of Leicester, UK.
| |
Collapse
|
45
|
Yuan X, Xie Z, Zou T. Recent advances in hypoxia-activated compounds for cancer diagnosis and treatment. Bioorg Chem 2024; 144:107161. [PMID: 38306826 DOI: 10.1016/j.bioorg.2024.107161] [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: 11/01/2023] [Revised: 12/28/2023] [Accepted: 01/27/2024] [Indexed: 02/04/2024]
Abstract
Hypoxia, as a prevalent feature of solid tumors, is correlated with tumorigenesis, proliferation, and invasion, playing an important role in mediating the drug resistance and affecting the cancer treatment outcomes. Due to the distinct oxygen levels between tumor and normal tissues, hypoxia-targeted therapy has attracted significant attention. The hypoxia-activated compounds mainly depend on reducible organic groups including azo, nitro, N-oxides, quinones and azide as well as some redox-active metal complex that are selectively converted into active species by the increased reduction potential under tumor hypoxia. In this review, we briefly summarized our current understanding on hypoxia-activated compounds with a particular highlight on the recently developed prodrugs and fluorescent probes for tumor treatment and diagnosis. We have also discussed the challenges and perspectives of small molecule-based hypoxia-activatable prodrug for future development.
Collapse
Affiliation(s)
- Xiaoyu Yuan
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhiying Xie
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Taotao Zou
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
46
|
Andrés CMC, Pérez de la Lastra JM, Juan CA, Plou FJ, Pérez-Lebeña E. Antioxidant Metabolism Pathways in Vitamins, Polyphenols, and Selenium: Parallels and Divergences. Int J Mol Sci 2024; 25:2600. [PMID: 38473850 DOI: 10.3390/ijms25052600] [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: 11/26/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Free radicals (FRs) are unstable molecules that cause reactive stress (RS), an imbalance between reactive oxygen and nitrogen species in the body and its ability to neutralize them. These species are generated by both internal and external factors and can damage cellular lipids, proteins, and DNA. Antioxidants prevent or slow down the oxidation process by interrupting the transfer of electrons between substances and reactive agents. This is particularly important at the cellular level because oxidation reactions lead to the formation of FR and contribute to various diseases. As we age, RS accumulates and leads to organ dysfunction and age-related disorders. Polyphenols; vitamins A, C, and E; and selenoproteins possess antioxidant properties and may have a role in preventing and treating certain human diseases associated with RS. In this review, we explore the current evidence on the potential benefits of dietary supplementation and investigate the intricate connection between SIRT1, a crucial regulator of aging and longevity; the transcription factor NRF2; and polyphenols, vitamins, and selenium. Finally, we discuss the positive effects of antioxidant molecules, such as reducing RS, and their potential in slowing down several diseases.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
| | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén, 7, 47011 Valladolid, Spain
| | - Francisco J Plou
- Institute of Catalysis and Petrochemistry, CSIC-Spanish Research Council, 28049 Madrid, Spain
| | | |
Collapse
|
47
|
Gupta N, Curcic M, Srivastava SK. Proguanil Suppresses Breast Tumor Growth In Vitro and In Vivo by Inducing Apoptosis via Mitochondrial Dysfunction. Cancers (Basel) 2024; 16:872. [PMID: 38473234 DOI: 10.3390/cancers16050872] [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: 11/27/2023] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Breast cancer, ranking as the second leading cause of female cancer-related deaths in the U.S., demands the exploration of innovative treatments. Repurposing FDA-approved drugs emerges as an expedited and cost-effective strategy. Our study centered on proguanil, an antimalarial drug, reveals notable anti-proliferative effects on diverse breast cancer cell lines, including those derived from patients. Proguanil-induced apoptosis was associated with a substantial increase in reactive oxygen species (ROS) production, leading to reduced mitochondrial membrane potential, respiration, and ATP production. Proguanil treatment upregulated apoptotic markers (Bax, p-H2AX, cleaved-caspase 3, 9, cleaved PARP) and downregulated anti-apoptotic proteins (bcl-2, survivin) in breast cancer cell lines. In female Balb/c mice implanted with 4T1 breast tumors, daily oral administration of 20 mg/kg proguanil suppressed tumor enlargement by 55%. Western blot analyses of proguanil-treated tumors supported the in vitro findings, demonstrating increased levels of p-H2AX, Bax, c-PARP, and c-caspase3 as compared to controls. Our results collectively highlight proguanil's anticancer efficacy in vitro and in vivo in breast cancer, prompting further consideration for clinical investigations.
Collapse
Affiliation(s)
- Nehal Gupta
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Marina Curcic
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Abilene, TX 79601, USA
| | - Sanjay K Srivastava
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Abilene, TX 79601, USA
- Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, 1718 Pine Street, Abilene, TX 79601, USA
| |
Collapse
|
48
|
Abdallah R, Shaito AA, Badran A, Baydoun S, Sobeh M, Ouchari W, Sahri N, Eid AH, Mesmar JE, Baydoun E. Fractionation and phytochemical composition of an ethanolic extract of Ziziphus nummularia leaves: antioxidant and anticancerous properties in human triple negative breast cancer cells. Front Pharmacol 2024; 15:1331843. [PMID: 38405665 PMCID: PMC10885810 DOI: 10.3389/fphar.2024.1331843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/09/2024] [Indexed: 02/27/2024] Open
Abstract
Natural products have long been utilized in traditional medicine as remedies to improve health and treat illnesses, and have had a key role in modern drug discovery. Recently, there has been a revived interest in the search for bioactives from natural sources as alternative or complementary modalities to synthetic medicines; especially for cancer treatment, which incidence and mortality rates are on the rise worldwide. Ziziphus nummularia has been widely used in traditional medicine for the treatment of various diseases. Its traditional uses and numerous ethnopharmacological properties may be attributed to its richness in bioactive metabolites. However, its phytochemical composition or chemopreventive effects against the aggressive triple-negative breast cancer (TNBC) are still poorly explored. Here, phytochemical composition of an ethanolic extract of Z. nummularia leaves (ZNE) and its chromatographically isolated fractions was identified both qualitatively by spectrophotometric assays and analytically by HPLC-PDA-MS/MS. The anti-proliferative effects of ZNE were tested in several cancer cell lines, but we focused on its anti-TNBC effects since they were not explored yet. The anti-cancerous potential of ZNE and its fractions was tested in vitro in MDA-MB-231, a TNBC cell line. Results showed that ZNE and its Fraction 6 (F6) reduced the viability of MDA-MB-231 cells. F6 decreased MDA-MB-231 viability more than crude ZNE or its other fractions. ZNE and F6 are rich in phytochemicals and HPLC-PDA-MS/MS analysis identified several metabolites that were previously reported to have anti-cancerous effects. Both ZNE and F6 showed potent antioxidant capacity in the DPPH assay, but promoted reactive oxygen species (ROS) production in MDA-MB-231 cells; an effect which was blunted by the antioxidant N-acetyl cysteine (NAC). NAC also blunted ZNE- and F6-induced reduction in TNBC cell viability. We also demonstrated that ZNE and F6 induced an arrest of the cell cycle, and triggered apoptosis- and autophagy-mediated cell death. ZNE and F6 inhibited metastasis-related cellular processes by modifying cell migration, invasion, and adhesion. Taken together, our findings reveal that Z. nummularia is rich in phytochemicals that can attenuate the malignant phenotype of TNBC and may offer innovative avenues for the discovery of new drug leads for treatment of TNBC and other cancers.
Collapse
Affiliation(s)
- Rola Abdallah
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Abdullah A. Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, and College of Medicine, Qatar University, Doha, Qatar
| | - Adnan Badran
- Department of Nutrition, University of Petra, Amman, Jordan
| | - Serine Baydoun
- Breast Imaging Section, Imaging Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Mansour Sobeh
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Wafae Ouchari
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Nihad Sahri
- Agrobiosciences Program, College for Agriculture and Environmental Science, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Elias Baydoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
49
|
Rauf A, Khalil AA, Awadallah S, Khan SA, Abu‐Izneid T, Kamran M, Hemeg HA, Mubarak MS, Khalid A, Wilairatana P. Reactive oxygen species in biological systems: Pathways, associated diseases, and potential inhibitors-A review. Food Sci Nutr 2024; 12:675-693. [PMID: 38370049 PMCID: PMC10867483 DOI: 10.1002/fsn3.3784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 02/20/2024] Open
Abstract
Reactive oxygen species (ROS) are produced under normal physiological conditions and may have beneficial and harmful effects on biological systems. ROS are involved in many physiological processes such as differentiation, proliferation, necrosis, autophagy, and apoptosis by acting as signaling molecules or regulators of transcription factors. In this case, maintaining proper cellular ROS levels is known as redox homeostasis. Oxidative stress occurs because of the imbalance between the production of ROS and antioxidant defenses. Sources of ROS include the mitochondria, auto-oxidation of glucose, and enzymatic pathways such as nicotinamide adenine dinucleotide phosphate reduced (NAD[P]H) oxidase. The possible ROS pathways are NF-κB, MAPKs, PI3K-Akt, and the Keap1-Nrf2-ARE signaling pathway. This review covers the literature pertaining to the possible ROS pathways and strategies to inhibit them. Additionally, this review summarizes the literature related to finding ROS inhibitors.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of ChemistryUniversity of SwabiAnbarPakistan
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahoreLahorePakistan
| | - Samir Awadallah
- Department of Medical Lab Sciences, Faculty of Allied Medical SciencesZarqa UniversityZarqaJordan
| | - Shahid Ali Khan
- Department of Chemistry, School of Natural SciencesNational University of Science and Technology (NUST)IslamabadPakistan
| | - Tareq Abu‐Izneid
- Pharmaceutical Sciences, College of PharmacyAl Ain UniversityAl Ain, Abu DhabiUAE
| | - Muhammad Kamran
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Hassan A. Hemeg
- Department of Medical Laboratory Technology, College of Applied Medical SciencesTaibah UniversityAl‐Medinah Al‐MonawaraSaudi Arabia
| | | | - Ahood Khalid
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahoreLahorePakistan
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical MedicineMahidol UniversityBangkokThailand
| |
Collapse
|
50
|
Chi T, Sang T, Wang Y, Ye Z. Cleavage and Noncleavage Chemistry in Reactive Oxygen Species (ROS)-Responsive Materials for Smart Drug Delivery. Bioconjug Chem 2024; 35:1-21. [PMID: 38118277 DOI: 10.1021/acs.bioconjchem.3c00476] [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: 12/22/2023]
Abstract
The design and development of advanced drug delivery systems targeting reactive oxygen species (ROS) have gained significant interest in recent years for treating various diseases, including cancer, psychiatric diseases, cardiovascular diseases, neurological diseases, metabolic diseases, and chronic inflammations. Integrating specific chemical bonds capable of effectively responding to ROS and triggering drug release into the delivery system is crucial. In this Review, we discuss commonly used conjugation linkers (chemical bonds) and categorize them into two groups: cleavable linkers and noncleavable linkers. Our goal is to clarify their unique drug release mechanisms from a chemical perspective and provide practical organic synthesis approaches for their efficient production. We showcase numerous significant examples to demonstrate their synthesis routes and diverse applications. Ultimately, we strive to present a comprehensive overview of cleavage and noncleavage chemistry, offering insights into the development of smart drug delivery systems that respond to ROS.
Collapse
Affiliation(s)
- Teng Chi
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ting Sang
- School of Stomatology of Nanchang University & Jiangxi Province Clinical Research Center for Oral Diseases & The Key Laboratory of Oral Biomedicine, Nanchang 330006, China
| | - Yanjing Wang
- Department of Chemistry, Washington University, St. Louis, Missouri 63130, United States
| | - Zhou Ye
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong S.A.R. 999077, China
| |
Collapse
|