1
|
Abusara OH, Hammad AM, Debas R, Al-Shalabi E, Waleed M, Scott Hall F. The inflammation and oxidative status of rat lung tissue following smoke/vapor exposure via E-cigarette, cigarette, and waterpipe. Gene 2025; 935:149066. [PMID: 39491601 DOI: 10.1016/j.gene.2024.149066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Tobacco smoking is a major worldwide health issue that contributes to millions of deaths annually. Electronic cigarettes (E-cigarettes) are also harmful. Smoke/vapor from E-cigarettes and tobacco products consists of free radicals and other toxic substances. Tissue damage in smokers, such as lungs, is highly observed and is linked to oxidative damage and inflammation. METHODS The inflammation and oxidative status of rat lung tissues was examined following whole-body smoke/vapor exposure via E-cigarette, cigarette, and waterpipe for 2 h daily, 5 days per week for 8 weeks. RESULTS Lung tissue damage was higher in cigarettes and waterpipe groups compared to the E-cigarette group. Collectively, there was a significant increase (p < 0.05) in the mRNA expression of pro-inflammatory mediators (TNF-α, NF-κB, IL-1β) with the exception of IL-1β in the E-cigarettes group. As for the anti-inflammatory mediators (Nrf2 and IL-10), a significant reduction (p < 0.05) of mRNA expression was observed with the exception of Nrf2 in the E-cigarette group. As for IL-6, there was a significant increase in its mRNA expression (p < 0.05) in the cigarette and waterpipe groups. There was also a significant decrease (p < 0.05) in the antioxidant activity of all antioxidants tested (GPx, SOD, and CAT) in all groups with the exception of SOD in the cigarette group. CONCLUSION Smoke/vapor administered via E-cigarette, cigarette, and waterpipe elicits inflammation and oxidative stress in rat lungs that is accompanied by histopathological changes.
Collapse
Affiliation(s)
- Osama H Abusara
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Alaa M Hammad
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan.
| | - Rasha Debas
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Eveen Al-Shalabi
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Mohammed Waleed
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
2
|
Wang B, Huang Y, Cheng X, Song J, Wang Q, Zhu Y, Zhang L, Liu G. Transduction of jellyfish superoxide dismutase mediated by TAT peptide ameliorates H 2O 2-induced oxidative stress in HaCaT cells. Sci Rep 2024; 14:31037. [PMID: 39730660 DOI: 10.1038/s41598-024-82261-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/03/2024] [Indexed: 12/29/2024] Open
Abstract
Superoxide dismutase (SOD) plays important roles in the balance of oxidation and antioxidation in body mostly by scavenging superoxide anion free radicals (O2.-). Previously, we reported a novel Cu/Zn SOD from jellyfish Cyanea capillata, named CcSOD1, which exhibited excellent SOD activity and high stability. TAT peptide is a common type of cell penetrating peptides (CPPs) that efficiently deliver extracellular biomacromolecules into cytoplasm. In this study, we constructed a recombinant expression vector that combined the coding sequences of TAT peptide and CcSOD1, and then obtained sufficient and high-purity TAT-CcSOD1 fusion protein. Compared with some reported SODs/CPP-SODs, TAT-CcSOD1 possessed stronger tolerance to heat and acid-base environment. TAT-CcSOD1 efficiently penetrated cell membrane and significantly enhanced the O2.- scavenging ability in cells, and attenuated H2O2-induced cytotoxicity and NO generation in HaCaT cells. This study serves as a critical step forward for the application of TAT-CcSOD1 as a potential protective/therapeutic agent against oxidative stress-related conditions in the future.
Collapse
Affiliation(s)
- Bo Wang
- Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China
- Naval Special Medical Center, Naval Medical University, Shanghai, 200052, China
| | - Yichao Huang
- Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China
| | - Xi Cheng
- Department of Pharmacy, Tianyou Hospital, Tongji University, Shanghai, China
| | - Juxingsi Song
- Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China
| | - Qianqian Wang
- Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China
| | - Yuanjie Zhu
- Naval Special Medical Center, Naval Medical University, Shanghai, 200052, China.
| | - Liming Zhang
- Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China.
| | - Guoyan Liu
- Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
3
|
Xu G, Yu J, Lyu J, Zhan M, Xu J, Huang M, Zhao R, Li Y, Zhu J, Feng J, Tan S, Ran P, Su Z, Liu X, Zhao J, Zhang H, Xu C, Chang J, Hou Y, Ding C. Proteogenomic Landscape of Breast Ductal Carcinoma Reveals Tumor Progression Characteristics and Therapeutic Targets. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401041. [PMID: 39418072 PMCID: PMC11633542 DOI: 10.1002/advs.202401041] [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: 01/28/2024] [Revised: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Multi-omics studies of breast ductal carcinoma (BRDC) have advanced the understanding of the disease's biology and accelerated targeted therapies. However, the temporal order of a series of biological events in the progression of BRDC is still poorly understood. A comprehensive proteogenomic analysis of 224 samples from 168 patients with malignant and benign breast diseases is carried out. Proteogenomic analysis reveals the characteristics of linear multi-step progression of BRDC, such as tumor protein P53 (TP53) mutation-associated estrogen receptor 1 (ESR1) overexpression is involved in the transition from ductal hyperplasia (DH) to ductal carcinoma in situ (DCIS). 6q21 amplification-associated nuclear receptor subfamily 3 group C member 1 (NR3C1) overexpression helps DCIS_Pure (pure DCIS, no histologic evidence of invasion) cells avoid immune destruction. The T-cell lymphoma invasion and metastasis 1, androgen receptor, and aldo-keto reductase family 1 member C1 (TIAM1-AR-AKR1C1) axis promotes cell invasion and migration in DCIS_adjIDC (DCIS regions of invasive cancers). In addition, AKR1C1 is identified as a potential therapeutic target and demonstrated the inhibitory effect of aspirin and dydrogesterone as its inhibitors on tumor cells. The integrative multi-omics analysis helps to understand the progression of BRDC and provides an opportunity to treat BRDC in different stages.
Collapse
Affiliation(s)
- Ganfei Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Juan Yu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jiacheng Lyu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Mengna Zhan
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jie Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Minjing Huang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Rui Zhao
- Institute for Developmental and Regenerative Cardiovascular MedicineMOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
| | - Yan Li
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jiajun Zhu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jinwen Feng
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Subei Tan
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Peng Ran
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Zhenghua Su
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Xinhua Liu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jianyuan Zhao
- Institute for Developmental and Regenerative Cardiovascular MedicineMOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
| | - Hongwei Zhang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Chen Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jun Chang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Yingyong Hou
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Chen Ding
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
- Departments of Cancer Research InstituteAffiliated Cancer Hospital of Xinjiang Medical UniversityXinjiang Key Laboratory of Translational Biomedical EngineeringUrumqi830000P. R. China
| |
Collapse
|
4
|
He X, Zhong L, Wang N, Zhao B, Wang Y, Wu X, Zheng C, Ruan Y, Hou J, Luo Y, Yin Y, He Y, Xiang AP, Wang J. Gastric Cancer Actively Remodels Mechanical Microenvironment to Promote Chemotherapy Resistance via MSCs-Mediated Mitochondrial Transfer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404994. [PMID: 39392399 DOI: 10.1002/advs.202404994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/03/2024] [Indexed: 10/12/2024]
Abstract
Chemotherapy resistance is the main reason of treatment failure in gastric cancer (GC). However, the mechanism of oxaliplatin (OXA) resistance remains unclear. Here, we demonstrate that extracellular mechanical signaling plays crucial roles in OXA resistance within GC. We selected OXA-resistant GC patients and analyzed tumor tissues by single-cell sequencing, and found that the mitochondrial content of GC cells increased in a biosynthesis-independent manner. Moreover, we found that the increased mitochondria of GC cells were mainly derived from mesenchymal stromal cells (MSCs), which could repair the mitochondrial function and reduce the levels of mitophagy in GC cells, thus leading to OXA resistance. Furthermore, we investigated the underlying mechanism and found that mitochondrial transfer was mediated by mechanical signals of the extracellular matrix (ECM). After OXA administration, GC cells actively secreted ECM in the tumor microenvironment (TEM), increasing matrix stiffness of the tumor tissues, which promoted mitochondria to transfer from MSCs to GC cells via microvesicles (MVs). Meanwhile, inhibiting the mechanical-related RhoA/ROCK1 pathway could alleviate OXA resistance in GC cells. In summary, these results indicate that matrix stiffness could be used as an indicator to identify chemotherapy resistance, and targeting mechanical-related pathway could effectively alleviate OXA resistance and improve therapeutic efficacy.
Collapse
Affiliation(s)
- Xin He
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Li Zhong
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Nan Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Baiwei Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yannan Wang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinxiang Wu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Changyu Zheng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Yueheng Ruan
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Jianfeng Hou
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yusheng Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yuehan Yin
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yulong He
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiancheng Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
5
|
van Boxel J, Khargi RRJ, Nijmeijer SM, Heinzelmann MT, Pereira DDC, Lamoree MH, van Duursen MBM. Effects of polystyrene micro- and nanoplastics on androgen- and estrogen receptor activity and steroidogenesis in vitro. Toxicol In Vitro 2024; 101:105938. [PMID: 39243830 DOI: 10.1016/j.tiv.2024.105938] [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: 07/12/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
While many plastic additives show endocrine disrupting properties, this has not been studied for micro- and nanoplastics (MNPs) particles despite their ubiquitous presence in humans. The objective of this study was to determine the effects of various sizes and concentrations of polystyrene (PS)-MNPs (50-10,000 nm, 0.01-100 μg/mL) on estrogen- and androgen receptor (ER and AR) activity and steroidogenesis in vitro. Fluorescent (F)PS-MNPs of ≤1000 nm were internalized in VM7 and H295R cells and FPS-MNPs ≤200 nm in AR-ecoscreen cells. H295R cells displayed the highest uptake and particles were closer to the nucleus than other cell types. None of the sizes and concentrations PS-MNPs tested affected ER or AR activity. In H295R cells, PS-MNPs caused some statistically significant changes in hormone levels, though these showed no apparent concentration or size-dependent patterns. Additionally, PS-MNPs caused a decrease in estriol (E3) with a maximum of 37.5 % (100 μg/mL, 50 nm) and an increase in gene expression of oxidative stress markers GPX1 (1.26-fold) and SOD1 (1.23-fold). Taken together, our data show limited endocrine-disrupting properties of PS-MNPs in vitro. Nevertheless the importance of E3 in the placenta warrants further studies in the potential effects of MNPs during pregnancy.
Collapse
Affiliation(s)
- Jeske van Boxel
- Amsterdam Institute for Life and Environment, section Environmental Health and Toxicology, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands.
| | - Rani R J Khargi
- Amsterdam Institute for Life and Environment, section Environmental Health and Toxicology, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Sandra M Nijmeijer
- Amsterdam Institute for Life and Environment, section Environmental Health and Toxicology, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Manuel T Heinzelmann
- Amsterdam Institute for Life and Environment, section Chemistry for Environment and Health, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Daniel Da Costa Pereira
- Division of Molecular and Computational Toxicology, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Marja H Lamoree
- Amsterdam Institute for Life and Environment, section Chemistry for Environment and Health, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Majorie B M van Duursen
- Amsterdam Institute for Life and Environment, section Environmental Health and Toxicology, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| |
Collapse
|
6
|
Besaratinia A, Caliri AW, Tommasi S. The interplay of DNA damage and repair, gene expression, and mutagenesis in mammalian cells during oxidative stress. Carcinogenesis 2024; 45:868-879. [PMID: 39023127 PMCID: PMC11584291 DOI: 10.1093/carcin/bgae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/13/2024] [Accepted: 07/16/2024] [Indexed: 07/20/2024] Open
Abstract
We investigated the interplay among oxidative DNA damage and repair, expression of genes encoding major base excision repair (BER) enzymes and bypass DNA polymerases, and mutagenesis in mammalian cells. Primary mouse embryonic fibroblasts were challenged with oxidative stress induced by methylene blue plus visible light, and formation and repair of DNA damage, changes in gene expression, and mutagenesis were determined at increasing intervals posttreatment (0-192 hours). Significant formation of oxidative DNA damage together with upregulation of Ogg1, Polβ, and Polκ, and no changes in Mutyh and Nudt1 expression were found in treated cells. There was a distinct interconnection between Ogg1 and Polβ expression and DNA damage formation and repair whereby changes in expression of these two genes were proportionate to the levels of oxidative DNA damage, once a 3-plus hour lag time passed (P < .05). Equally notable was the matching pattern of Polκ expression and kinetics of oxidative DNA damage and repair (P < .05). The DNA damage and gene expression data were remarkably consistent with mutagenicity data in the treated cells; the induced mutation spectrum is indicative of erroneous bypass of oxidized DNA bases and incorporation of oxidized deoxynucleoside triphosphates during replication of the genomic DNA. Our findings support follow-up functional studies to elucidate how oxidation of DNA bases and the nucleotide pool, overexpression of Polκ, delayed upregulation of Ogg1 and Polβ, and inadequate expression of Nudt1 and Mutyh collectively affect mutagenesis consequent to oxidative stress.
Collapse
Affiliation(s)
- Ahmad Besaratinia
- Department of Population & Public Health Sciences, USC Keck School of Medicine, University of Southern California, M/C 9603, Los Angeles, CA 90033, United States
| | - Andrew W Caliri
- Department of Population & Public Health Sciences, USC Keck School of Medicine, University of Southern California, M/C 9603, Los Angeles, CA 90033, United States
| | - Stella Tommasi
- Department of Population & Public Health Sciences, USC Keck School of Medicine, University of Southern California, M/C 9603, Los Angeles, CA 90033, United States
| |
Collapse
|
7
|
Palafox MA, Belskaya NP, Todorov L, Hristova-Avakumova N, Kostova IP. Molecular properties of a triazole-Ce(III) complex with antioxidant activity: structure, spectroscopy, and relationships with related derivatives. Influence of the ligands in the complex. Front Chem 2024; 12:1450106. [PMID: 39569014 PMCID: PMC11576285 DOI: 10.3389/fchem.2024.1450106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 10/04/2024] [Indexed: 11/22/2024] Open
Abstract
A novel Ce(III) complex with the triazole ligand 2b, which presents four H-bonded sites with amino acids of the MMP-2 receptor, was synthesized. The experimental IR and Raman spectra of this Ce(III) complex were well-interpreted based on their comparison to the theoretical scaled spectra using the scaling equations determined by two procedures and four density functional theory (DFT) levels. Therefore, the structure predicted for the synthesized Ce(III) complex was clearly characterized and confirmed. The potential antioxidant action of this complex was compared with the analogous La(III) complex, and it was found that the coordination of ligand 2b with Ce(III) improves the ligand's ability to participate in single-electron transfer (SET), as observed in the ABTS·+ assay, and this complex seems to scavenge the stable radical much more actively compared to its La(III) counterpart. Additionally, interactions with potassium superoxide and sodium hypochlorite indicate a high pro-oxidant behavior of the complex. The effects of different ligands on the geometric parameters, atomic charges, and molecular properties of the Ce(III) complex were analyzed at four DFT levels, and several relationships were clearly established. These relationships can facilitate the selection of new ligands with improved properties in the design of novel lanthanide-triazole carboxylate complexes with promising biological activity. The ligand size increase in the complexes facilitates the electronic transfer of negative charge, and the low HOMO (highest occupied molecular orbital)-LUMO (lowest unoccupied molecular orbital) energy gap indicates a large reactivity and low energy for their excitation.
Collapse
Affiliation(s)
- M Alcolea Palafox
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - Nataliya P Belskaya
- Department of Technology for Organic Synthesis, Ural Federal University, Yekaterinburg, Russia
| | - Lozan Todorov
- Department of Chemistry, Faculty of Pharmacy, Medical University - Sofia, Sofia, Bulgaria
| | - Nadya Hristova-Avakumova
- Department of Medical Physics and Biophysics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Irena P Kostova
- Department of Chemistry, Faculty of Pharmacy, Medical University - Sofia, Sofia, Bulgaria
| |
Collapse
|
8
|
Mao Y, Sha R, Sun Y, Wang Z, Huang J. Antioxidative and Cytoprotective Effects of Rosa Roxburghii and Metabolite Changes in Oxidative Stress-Induced HepG2 Cells Following Rosa Roxburghii Intervention. Foods 2024; 13:3520. [PMID: 39517304 PMCID: PMC11545593 DOI: 10.3390/foods13213520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
Rosa Roxburghii (RR), a traditional Chinese medicinal fruit, is rich in bioactive substances that make it a potential natural antioxidant resource. This research aimed to study the antioxidant properties of RR by in vitro experiments and through intracellular assessment in H2O2-induced HepG2 cells. A non-targeted metabolic analysis was conducted to indicate changes in intracellular and extracellular metabolites. Differential metabolites and metabolic pathways were explored using PCA, PLS-DA, and KEGG pathway analysis. The results showed that RR rich in bioactive substances exhibited a significant antioxidative property in vitro and intracellularly. This property may be achieved by scavenging free radicals, increasing the activity of catalase (CAT), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), and the levels of bicinchoninic acid (BCA) while reducing the reactive oxygen species (ROS) generation. This study identified 13 differential metabolites intracellularly and 7 extracellularly, among which the key differential metabolites included D-glucopyranose, D-mannose, fructose, citric acid, malic acid, cholesterol, and cholestenone. These key metabolites primarily regulated glucose-related metabolism, the citrate cycle, and the primary bile acid biosynthesis pathway in H2O2-induced HepG2 cells. These findings provide potential application evidence of RR in the development of natural resources for functional foods.
Collapse
Affiliation(s)
- Yangchen Mao
- Zhejiang Provincial Key Laboratory for Chemical & Biological Processing Technology of Farm Product, Hangzhou 310023, China; (Y.M.)
- Zhejiang Province Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Ruyi Sha
- Zhejiang Provincial Key Laboratory for Chemical & Biological Processing Technology of Farm Product, Hangzhou 310023, China; (Y.M.)
- Zhejiang Province Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Yuhao Sun
- Zhejiang Provincial Key Laboratory for Chemical & Biological Processing Technology of Farm Product, Hangzhou 310023, China; (Y.M.)
- Zhejiang Province Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Zhenzhen Wang
- Zhejiang Provincial Key Laboratory for Chemical & Biological Processing Technology of Farm Product, Hangzhou 310023, China; (Y.M.)
- Zhejiang Province Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Jun Huang
- Zhejiang Provincial Key Laboratory for Chemical & Biological Processing Technology of Farm Product, Hangzhou 310023, China; (Y.M.)
- Zhejiang Province Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| |
Collapse
|
9
|
Aliko V, Vasjari L, Istifli ES, Gjonaj G, Impellitteri F, Faggio C, Benedetti E, Zugaro S, Iannetta A, Perugini M. Molecular docking analysis and in vivo assessment of zinc oxide nanoparticle toxicity in zebrafish larvae. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 276:107112. [PMID: 39383782 DOI: 10.1016/j.aquatox.2024.107112] [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: 05/21/2024] [Revised: 08/12/2024] [Accepted: 09/29/2024] [Indexed: 10/11/2024]
Abstract
The zinc oxide nanoparticles (ZnO-NPs) being widely employed in several industries and consumer products, are raising concerns about their safety on aquatic biota and human health. This study aims to investigate the possible toxicological effects of ZnO-NPs through a combined in vivo and in silico approach. Zebrafish embryos were exposed to several ZnO-NPs concentrations and morphological alterations and lipid peroxidation (MDA) were investigated. Furthermore, molecular docking simulations were applied to study the intermolecular interactions of ZnO-NPs against critical embryonic proteins namely zebrafish hatching enzyme1 (ZHE1) as well as the superoxide dismutase (SOD1). Treatment with ZnO-NPs resulted in an increase in MDA concentration and a decrease in antioxidant enzyme levels. Besides a significant decrease in mRNA expression of key enzymes of ROS detoxification genes, a modulation of inflammatory genes with a low downregulation of tnf-α, and an upregulation of il-1β were observed. Docking study suggests that the delayed hatching and increased cellular oxidative stress in zebrafish embryos may occur through a synergistic mechanism based on the ZnO-NP-dependent inhibition of ZHE1 and SOD1 enzymes. The integration of in vivo assessments with in silico computational modeling provided a more comprehensive evaluation of potential physiological risks in zebrafish embryos associated with nanomaterial exposure.
Collapse
Affiliation(s)
- Valbona Aliko
- University of Tirana, Faculty of Natural Sciences, Department of Biology, Tirana, Albania
| | - Ledia Vasjari
- University of Tirana, Faculty of Natural Sciences, Department of Biology, Tirana, Albania
| | - Erman S Istifli
- University of Cukurova, Faculty of Science and Literature, Department of Biology, Adana, Turkey
| | - Grejsi Gjonaj
- University of Tirana, Faculty of Natural Sciences, Department of Biology, Tirana, Albania
| | | | - Caterina Faggio
- University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Messina, Italy; Department of Eco-sustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Silvana Zugaro
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annamaria Iannetta
- University of Teramo, Department of Bioscience and Agro-Food and Environmental Technology, Teramo, Italy.
| | - Monia Perugini
- University of Teramo, Department of Bioscience and Agro-Food and Environmental Technology, Teramo, Italy
| |
Collapse
|
10
|
Kado A, Moriya K, Inoue Y, Yanagimoto S, Tsutsumi T, Koike K, Fujishiro M. Decreased antioxidant-related superoxide dismutase 1 expression in peripheral immune cells indicates early ethanol exposure. Sci Rep 2024; 14:25091. [PMID: 39443615 PMCID: PMC11499712 DOI: 10.1038/s41598-024-76084-8] [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: 04/07/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
Alcohol consumption increases oxidative stress and imbalances in the antioxidant system, even with ethanol (EtOH) exposure at a young age. This study assessed changes in the antioxidant system following young EtOH exposure in peripheral immunity and measured sensitive indicators of heavy alcohol consumption. We used peripheral blood mononuclear cells (PBMCs) from 197 male university students without smoking habits to examine changes in antioxidant-related gene expression in vitro and in PBMCs. In vitro, the antioxidant system was impaired by EtOH. Next, we examined the expression of 84 antioxidant-related genes in the PBMCs of 162 young adults, among which the superoxide dismutase (SOD) 1 expression was most negatively correlated with alcohol consumption degree. The plasma SOD1 level had the highest area under the curve value (0.806) for heavy alcohol consumption. Our data demonstrated that a decreased SOD1 level is a sensitive indicator of an impaired antioxidant system and heavy alcohol consumption with early EtOH exposure.
Collapse
Affiliation(s)
- Akira Kado
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Division for Health Service Promotion, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kyoji Moriya
- Division of Infection Control and Prevention, Education Research Center, Tokyo Health Care University, 4-1-17 Higashigotanda, Shinagawa-ku, Tokyo, 141-8648, Japan.
- Department of Infection Control and Prevention, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Yukiko Inoue
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Division for Health Service Promotion, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shintaro Yanagimoto
- Division for Health Service Promotion, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takeya Tsutsumi
- Department of Infection Control and Prevention, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Gastroenterology, Kanto Central Hospital, 6-25-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8531, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
11
|
Godet I, Oza HH, Shi Y, Joe NS, Weinstein AG, Johnson J, Considine M, Talluri S, Zhang J, Xu R, Doctorman S, Mbulaiteye D, Stein-O'Brien G, Kagohara LT, Santa-Maria CA, Fertig EJ, Gilkes DM. Hypoxia induces ROS-resistant memory upon reoxygenation in vivo promoting metastasis in part via MUC1-C. Nat Commun 2024; 15:8416. [PMID: 39341835 PMCID: PMC11438863 DOI: 10.1038/s41467-024-51995-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Hypoxia occurs in 90% of solid tumors and is associated with metastasis and mortality. Breast cancer cells that experience intratumoral hypoxia are 5x more likely to develop lung metastasis in animal models. Using spatial transcriptomics, we determine that hypoxic cells localized in more oxygenated tumor regions (termed 'post-hypoxic') retain expression of hypoxia-inducible and NF-kB-regulated genes, even in the oxygen-rich bloodstream. This cellular response is reproduced in vitro under chronic hypoxic conditions followed by reoxygenation. A subset of genes remains increased in reoxygenated cells. MUC1/MUC1-C is upregulated by both HIF-1α and NF-kB-p65 during chronic hypoxia. Abrogating MUC1 decreases the expression of superoxide dismutase enzymes, causing reactive oxygen species (ROS) production and cell death. A hypoxia-dependent genetic deletion of MUC1, or MUC1-C inhibition by GO-203, increases ROS levels in circulating tumor cells (CTCs), reducing the extent of metastasis. High MUC1 expression in tumor biopsies is associated with recurrence, and MUC1+ CTCs have lower ROS levels than MUC1- CTCs in patient-derived xenograft models. This study demonstrates that therapeutically targeting MUC1-C reduces hypoxia-driven metastasis.
Collapse
Affiliation(s)
- Inês Godet
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, USA
| | - Harsh H Oza
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yi Shi
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Natalie S Joe
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alyssa G Weinstein
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Biochemistry and Molecular Biology Program, The Johns Hopkins University School of Public Health, Baltimore, MD, USA
| | - Jeanette Johnson
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Considine
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Swathi Talluri
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jingyuan Zhang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Biochemistry and Molecular Biology Program, The Johns Hopkins University School of Public Health, Baltimore, MD, USA
| | - Reid Xu
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Steven Doctorman
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Delma Mbulaiteye
- NIDDK STEP-UP Program, National Institutes of Health, Bethesda, USA
| | - Genevieve Stein-O'Brien
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luciane T Kagohara
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cesar A Santa-Maria
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elana J Fertig
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Daniele M Gilkes
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA.
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, USA.
- Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Panda B, Tripathy A, Patra S, Kullu B, Tabrez S, Jena M. Imperative connotation of SODs in cancer: Emerging targets and multifactorial role of action. IUBMB Life 2024; 76:592-613. [PMID: 38600696 DOI: 10.1002/iub.2821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024]
Abstract
Superoxide dismutase (SOD) is a crucial enzyme responsible for the redox homeostasis inside the cell. As a part of the antioxidant defense system, it plays a pivotal role in the dismutation of the superoxide radicals (O 2 - ) generated mainly by the oxidative phosphorylation, which would otherwise bring out the redox dysregulation, leading to higher reactive oxygen species (ROS) generation and, ultimately, cell transformation, and malignancy. Several studies have shown the involvement of ROS in a wide range of human cancers. As SOD is the key enzyme in regulating ROS, any change, such as a transcriptional change, epigenetic remodeling, functional alteration, and so forth, either activates the proto-oncogenes or aberrant signaling cascades, which results in cancer. Interestingly, in some cases, SODs act as tumor promoters instead of suppressors. Furthermore, SODs have also been known to switch their role during tumor progression. In this review, we have tried to give a comprehensive account of SODs multifactorial role in various human cancers so that SODs-based therapeutic strategies could be made to thwart cancers.
Collapse
Affiliation(s)
- Biswajit Panda
- Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Ankita Tripathy
- Post Graduate Department of Botany, Utkal University, Bhubaneswar, India
| | - Srimanta Patra
- Post Graduate Department of Botany, Berhampur University, Berhampur, India
| | - Bandana Kullu
- Post Graduate Department of Botany, Utkal University, Bhubaneswar, India
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mrutyunjay Jena
- Post Graduate Department of Botany, Berhampur University, Berhampur, India
| |
Collapse
|
13
|
König S, Strassheimer F, Brandner NI, Schröder JH, Urban H, Harwart LF, Hehlgans S, Steinbach JP, Ronellenfitsch MW, Luger AL. Superoxide dismutase 1 mediates adaptation to the tumor microenvironment of glioma cells via mammalian target of rapamycin complex 1. Cell Death Discov 2024; 10:379. [PMID: 39187509 PMCID: PMC11347576 DOI: 10.1038/s41420-024-02145-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024] Open
Abstract
In glioblastoma (GB) cells oxidative stress is induced by both, conditions of the tumor microenvironment as well as by therapeutic interventions. Upregulation of superoxide dismutase 1 (SOD1), a key enzyme for oxidative defense and downstream target of mammalian target of rapamycin complex 1 (mTORC1) is a candidate mechanism to sustain survival and proliferation of tumor cells. SOD1 was inhibited by shRNA mediated gene suppression, CRISPR/Cas9 knockout and pharmacological inhibition in human (primary) GB cells. SOD1 activity was determined by SOD1/2 activity assay. ROS levels, cell death and the NADPH/NADP-ratio were measured under normal and starvation conditions. To study the mTORC1-SOD1 axis, mTORC1 activated TSC2 knockdown cells (TSC2sh) were analyzed. Genetic and pharmacological inhibition of SOD1 correlated with decreased SOD1 activity, increased ROS and enhanced the sensitivity of glioma cells towards starvation- and hypoxia-induced cell death. This was accompanied by a decreased NADPH/NADP-ratio. Furthermore, combination therapy of SOD1 and mTORC1 inhibition partially rescued the protective effect of mTORC1 inhibitor monotherapy. SOD1 mediates adaptation of GB cells to stress conditions in the tumor microenvironment in a mTORC1-dependent manner. Moreover, SOD1 activation contributes to the cell death resistance conferred by mTORC1 inhibitors under hypoxic conditions.
Collapse
Affiliation(s)
- Sven König
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany
| | - Florian Strassheimer
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany
| | - Nadja I Brandner
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany
| | - Jan-Hendrik Schröder
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany
| | - Hans Urban
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany
| | - Leander F Harwart
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany
| | - Stephanie Hehlgans
- Goethe University Frankfurt, University Hospital, Department of Radiotherapy and Oncology, Frankfurt am Main, Germany
| | - Joachim P Steinbach
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany
| | - Michael W Ronellenfitsch
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany
| | - Anna-Luisa Luger
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany.
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany.
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany.
| |
Collapse
|
14
|
Kejík Z, Hajduch J, Abramenko N, Vellieux F, Veselá K, Fialová JL, Petrláková K, Kučnirová K, Kaplánek R, Tatar A, Skaličková M, Masařík M, Babula P, Dytrych P, Hoskovec D, Martásek P, Jakubek M. Cyanine dyes in the mitochondria-targeting photodynamic and photothermal therapy. Commun Chem 2024; 7:180. [PMID: 39138299 PMCID: PMC11322665 DOI: 10.1038/s42004-024-01256-6] [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: 02/08/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
Mitochondrial dysregulation plays a significant role in the carcinogenesis. On the other hand, its destabilization strongly represses the viability and metastatic potential of cancer cells. Photodynamic and photothermal therapies (PDT and PTT) target mitochondria effectively, providing innovative and non-invasive anticancer therapeutic modalities. Cyanine dyes, with strong mitochondrial selectivity, show significant potential in enhancing PDT and PTT. The potential and limitations of cyanine dyes for mitochondrial PDT and PTT are discussed, along with their applications in combination therapies, theranostic techniques, and optimal delivery systems. Additionally, novel approaches for sonodynamic therapy using photoactive cyanine dyes are presented, highlighting advances in cancer treatment.
Collapse
Affiliation(s)
- Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic.
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic.
| | - Jan Hajduch
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Nikita Abramenko
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Frédéric Vellieux
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | | | - Kateřina Petrláková
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00, Brno, Czech Republic
| | - Kateřina Kučnirová
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Ameneh Tatar
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Markéta Skaličková
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
| | - Michal Masařík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 121 08, Prague, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 121 08, Prague, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic.
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Prague, Czech Republic.
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455, 120 00, Prague, Czech Republic.
| |
Collapse
|
15
|
Zhang KH, Jiao L, Wang Y, Sun SC. Arf6 GTPase deficiency leads to porcine oocyte quality decline during aging. FASEB J 2024; 38:e23739. [PMID: 38884157 DOI: 10.1096/fj.202400893r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/18/2024]
Abstract
Arf6 is a member of ADP-ribosylation factor (Arf) family, which is widely implicated in the regulation of multiple physiological processes including endocytic recycling, cytoskeletal organization, and membrane trafficking during mitosis. In this study, we investigated the potential relationship between Arf6 and aging-related oocyte quality, and its roles on organelle rearrangement and cytoskeleton dynamics in porcine oocytes. Arf6 expressed in porcine oocytes throughout meiotic maturation, and it decreased in aged oocytes. Disruption of Arf6 led to the failure of cumulus expansion and polar body extrusion. Further analysis indicated that Arf6 modulated ac-tubulin for meiotic spindle organization and microtubule stability. Besides, Arf6 regulated cofilin phosphorylation and fascin for actin assembly, which further affected spindle migration, indicating the roles of Arf6 on cytoskeleton dynamics. Moreover, the lack of Arf6 activity caused the dysfunction of Golgi and ER for protein synthesis and signal transduction. Mitochondrial dysfunction was also observed in Arf6-deficient porcine oocytes, which was supported by the increased ROS level and abnormal membrane potential. In conclusion, our results reported that insufficient Arf6 was related to aging-induced oocyte quality decline through spindle organization, actin assembly, and organelle rearrangement in porcine oocytes.
Collapse
Affiliation(s)
- Kun-Huan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Le Jiao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yue Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
16
|
Zhou Y, Wang C, Nie Y, Wu L, Xu A. 2,4,6-trinitrotoluene causes mitochondrial toxicity in Caenorhabditis elegans by affecting electron transport. ENVIRONMENTAL RESEARCH 2024; 252:118820. [PMID: 38555093 DOI: 10.1016/j.envres.2024.118820] [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: 02/04/2024] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
As a typical energetic compound widely used in military activities, 2,4,6-trinitrotoluene (TNT) has attracted great attention in recent years due to its heavy pollution and wide distribution in and around the training facilities, firing ranges, and demolition sites. However, the subcellular targets and the underlying toxic mechanism of TNT remain largely unknown. In this study, we explored the toxic effects of TNT biological reduction on the mitochondrial function and homeostasis in Caenorhabditis elegans (C. elegans). With short-term exposure of L4 larvae, 10-1000 ng/mL TNT reduced mitochondrial membrane potential and adenosine triphosphate (ATP) content, which was associated with decreased expression of specific mitochondrial complex involving gas-1 and mev-1 genes. Using fluorescence-labeled transgenic nematodes, we found that fluorescence expression of sod-3 (muls84) and gst-4 (dvls19) was increased, suggesting that TNT disrupted the mitochondrial antioxidant defense system. Furthermore, 10 ng/mL TNT exposure increased the expression of the autophagy-related gene pink-1 and activated mitochondrial unfolded protein response (mt UPR), which was indicated by the increased expression of mitochondrial stress activated transcription factor atfs-1, ubiquitin-like protein ubl-5, and homeobox protein dve-1. Our findings demonstrated that TNT biological reduction caused mitochondrial dysfunction and the development of mt UPR protective stress responses, and provided a basis for determining the potential risks of energetic compounds to living organisms.
Collapse
Affiliation(s)
- Yanping Zhou
- Center of Free Electron Laser & High Magnetic Field, Anhui University, Hefei, 230601, PR China
| | - Chunyan Wang
- Center of Free Electron Laser & High Magnetic Field, Anhui University, Hefei, 230601, PR China
| | - Yaguang Nie
- Center of Free Electron Laser & High Magnetic Field, Anhui University, Hefei, 230601, PR China.
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - An Xu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory, HFIPS, Chinese Academy of Science, Anhui, Hefei, 230031, PR China.
| |
Collapse
|
17
|
Piazza CE, Mattos JJ, Lima D, Siebert MN, Zacchi FL, Dos Reis ÍMM, Ferrari FL, Balsanelli E, Toledo-Silva G, de Souza EM, Bainy ACD. Hepatic transcriptome, transcriptional effects and antioxidant responses in Poecilia vivipara exposed to sanitary sewage. MARINE POLLUTION BULLETIN 2024; 203:116426. [PMID: 38692005 DOI: 10.1016/j.marpolbul.2024.116426] [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: 11/23/2023] [Revised: 04/05/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Aquatic environments are subject to threats from multiple human activities, particularly through the release of untreated sanitary sewage into the coastal environments. These effluents contain a large group of natural or synthetic compounds referred to as emerging contaminants. Monitoring the types and quantities of toxic substances in the environment, especially complex mixtures, is an exhausting and challenging task. Integrative effect-based tools, such as biomarkers, are recommended for environmental quality monitoring programs. In this study, fish Poecilia vivipara were exposed for 24 and 96 h to raw untreated sewage diluted 33 % (v/v) in order to identify hepatic genes to be used as molecular biomarkers. Through a de novo hepatic transcriptome assembly, using Illumina MiSeq, 54,285 sequences were assembled creating a reference transcriptome for this guppy species. Transcripts involved in biotransformation systems, antioxidant defenses, ABC transporters, nuclear and xenobiotic receptors were identified and evaluated by qPCR. Sanitary sewage induced transcriptional changes in AhR, PXR, CYP2K1, CYP3A30, NQO1, UGT1A1, GSTa3, GSTmu, ST1C1, SOD, ABCC1 and SOX9 genes from liver of fish, particularly after 96 h of exposure. Changes in hepatic enzyme activities were also observed. The enzymes showed differences in fish exposed to both periods, while in the gills there was a prevalence of significant results after 96 h. The observed differences were associated to gender and/or to sewage exposure. The obtained results support the use of P. vivipara as sentinel and model organism for ecotoxicological studies and evidence the importance of understanding the differential responses associated to gender.
Collapse
Affiliation(s)
- Clei Endrigo Piazza
- Laboratory of Biomarkers of Aquatic Contamination and Immunochemistry, Department of Biochemistry, Federal University of Santa Catarina, UFSC, Florianópolis, SC, Brazil
| | - Jacó Joaquim Mattos
- Aquaculture Pathology Research, NEPAQ, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Daína Lima
- Laboratory of Biomarkers of Aquatic Contamination and Immunochemistry, Department of Biochemistry, Federal University of Santa Catarina, UFSC, Florianópolis, SC, Brazil
| | - Marília Nardelli Siebert
- Laboratory of Biomarkers of Aquatic Contamination and Immunochemistry, Department of Biochemistry, Federal University of Santa Catarina, UFSC, Florianópolis, SC, Brazil
| | - Flávia Lucena Zacchi
- Laboratory of Biomarkers of Aquatic Contamination and Immunochemistry, Department of Biochemistry, Federal University of Santa Catarina, UFSC, Florianópolis, SC, Brazil
| | - Ísis Mayna Martins Dos Reis
- Laboratory of Biomarkers of Aquatic Contamination and Immunochemistry, Department of Biochemistry, Federal University of Santa Catarina, UFSC, Florianópolis, SC, Brazil
| | - Fernanda Luiza Ferrari
- Bioinformatics Laboratory, Cell Biology, Embriology and Genetics Department, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Eduardo Balsanelli
- Department of Biochemistry, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Guilherme Toledo-Silva
- Bioinformatics Laboratory, Cell Biology, Embriology and Genetics Department, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | | | - Afonso Celso Dias Bainy
- Laboratory of Biomarkers of Aquatic Contamination and Immunochemistry, Department of Biochemistry, Federal University of Santa Catarina, UFSC, Florianópolis, SC, Brazil.
| |
Collapse
|
18
|
Kim MB, Park SM, Lim GH, Oh YH, Seo KW, Youn HY. Neuroprotective and immunomodulatory effects of superoxide dismutase on SH-SY5Y neuroblastoma cells and RAW264.7 macrophages. PLoS One 2024; 19:e0303136. [PMID: 38743689 PMCID: PMC11093368 DOI: 10.1371/journal.pone.0303136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/19/2024] [Indexed: 05/16/2024] Open
Abstract
Superoxide dismutase (SOD) is an antioxidant enzyme that protects the body from free radicals. It has both antioxidant and immunomodulatory properties, inducing macrophage polarization from M1 to M2. Macrophages, key mediators of the innate immune response, are divided into the M1 (pro-inflammatory) and M2 (anti-inflammatory) subtypes. In this study, we aimed to assess the antioxidant and neuroprotective effects of SOD on nerve cells and its immunomodulatory effects on macrophages. We observed that SOD inhibited the accumulation of reactive oxygen species and enhanced the viability of H2O2-treated nerve cells. Furthermore, SOD reduced the degree of necrosis in nerve cells treated with the conditioned medium from macrophages, which induced inflammation. In addition, SOD promoted the M1 to M2 transition of macrophages. Our findings suggest that SOD protects nerve cells and regulates immune responses.
Collapse
Affiliation(s)
- Moon-Beom Kim
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Su-Min Park
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ga-Hyun Lim
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yong-Hun Oh
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Won Seo
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hwa-Young Youn
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
19
|
Ramos-Acosta C, Huerta-Pantoja L, Salazar-Hidalgo ME, Mayol E, Jiménez-Vega S, García-Peña P, Jordi-Cruz J, Baquero C, Porras A, Íñigo-Rodríguez B, Benavente CM, López-Pastor AR, Gómez-Delgado I, Urcelay E, Candel FJ, Anguita E. Tigecycline Opposes Bortezomib Effect on Myeloma Cells Decreasing Mitochondrial Reactive Oxygen Species Production. Int J Mol Sci 2024; 25:4887. [PMID: 38732105 PMCID: PMC11084384 DOI: 10.3390/ijms25094887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Multiple myeloma is an incurable plasma cell malignancy. Most patients end up relapsing and developing resistance to antineoplastic drugs, like bortezomib. Antibiotic tigecycline has activity against myeloma. This study analyzed tigecycline and bortezomib combination on cell lines and plasma cells from myeloma patients. Apoptosis, autophagic vesicles, mitochondrial mass, mitochondrial superoxide, cell cycle, and hydrogen peroxide were studied by flow cytometry. In addition, mitochondrial antioxidants and electron transport chain complexes were quantified by reverse transcription real-time PCR (RT-qPCR) or western blot. Cell metabolism and mitochondrial activity were characterized by Seahorse and RT-qPCR. We found that the addition of tigecycline to bortezomib reduces apoptosis in proportion to tigecycline concentration. Supporting this, the combination of both drugs counteracts bortezomib in vitro individual effects on the cell cycle, reduces autophagy and mitophagy markers, and reverts bortezomib-induced increase in mitochondrial superoxide. Changes in mitochondrial homeostasis and MYC upregulation may account for some of these findings. These data not only advise to avoid considering tigecycline and bortezomib combination for treating myeloma, but caution on the potential adverse impact of treating infections with this antibiotic in myeloma patients under bortezomib treatment.
Collapse
Affiliation(s)
- Carlos Ramos-Acosta
- Department of Medicine, Medical School, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040 Madrid, Spain (E.M.); (S.J.-V.); (J.J.-C.); (C.M.B.); (F.J.C.)
- Hematology Department, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Profesor Martín Lagos s/n, 28040 Madrid, Spain (B.Í.-R.)
| | - Laura Huerta-Pantoja
- Department of Medicine, Medical School, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040 Madrid, Spain (E.M.); (S.J.-V.); (J.J.-C.); (C.M.B.); (F.J.C.)
- Hematology Department, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Profesor Martín Lagos s/n, 28040 Madrid, Spain (B.Í.-R.)
| | - Milton Eduardo Salazar-Hidalgo
- Hematology Department, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Profesor Martín Lagos s/n, 28040 Madrid, Spain (B.Í.-R.)
| | - Elsa Mayol
- Department of Medicine, Medical School, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040 Madrid, Spain (E.M.); (S.J.-V.); (J.J.-C.); (C.M.B.); (F.J.C.)
- Hematology Department, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Profesor Martín Lagos s/n, 28040 Madrid, Spain (B.Í.-R.)
| | - Selene Jiménez-Vega
- Department of Medicine, Medical School, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040 Madrid, Spain (E.M.); (S.J.-V.); (J.J.-C.); (C.M.B.); (F.J.C.)
- Hematology Department, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Profesor Martín Lagos s/n, 28040 Madrid, Spain (B.Í.-R.)
| | - Pablo García-Peña
- Department of Medicine, Medical School, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040 Madrid, Spain (E.M.); (S.J.-V.); (J.J.-C.); (C.M.B.); (F.J.C.)
- Hematology Department, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Profesor Martín Lagos s/n, 28040 Madrid, Spain (B.Í.-R.)
| | - Jenifeer Jordi-Cruz
- Department of Medicine, Medical School, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040 Madrid, Spain (E.M.); (S.J.-V.); (J.J.-C.); (C.M.B.); (F.J.C.)
- Hematology Department, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Profesor Martín Lagos s/n, 28040 Madrid, Spain (B.Í.-R.)
| | - Cristina Baquero
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (C.B.); (A.P.)
| | - Almudena Porras
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (C.B.); (A.P.)
| | - Belén Íñigo-Rodríguez
- Hematology Department, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Profesor Martín Lagos s/n, 28040 Madrid, Spain (B.Í.-R.)
| | - Celina M. Benavente
- Department of Medicine, Medical School, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040 Madrid, Spain (E.M.); (S.J.-V.); (J.J.-C.); (C.M.B.); (F.J.C.)
- Hematology Department, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Profesor Martín Lagos s/n, 28040 Madrid, Spain (B.Í.-R.)
| | - Andrea R. López-Pastor
- Laboratory of Genetics and Molecular Bases of Complex Diseases, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (A.R.L.-P.); (I.G.-D.); (E.U.)
- Networks for Cooperative Research in Health Results (RICORS, REI), 28089 Madrid, Spain
| | - Irene Gómez-Delgado
- Laboratory of Genetics and Molecular Bases of Complex Diseases, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (A.R.L.-P.); (I.G.-D.); (E.U.)
- Networks for Cooperative Research in Health Results (RICORS, REI), 28089 Madrid, Spain
| | - Elena Urcelay
- Laboratory of Genetics and Molecular Bases of Complex Diseases, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (A.R.L.-P.); (I.G.-D.); (E.U.)
- Networks for Cooperative Research in Health Results (RICORS, REI), 28089 Madrid, Spain
| | - Francisco Javier Candel
- Department of Medicine, Medical School, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040 Madrid, Spain (E.M.); (S.J.-V.); (J.J.-C.); (C.M.B.); (F.J.C.)
- Clinical Microbiology & Infectious Diseases, Transplant Coordination, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Eduardo Anguita
- Department of Medicine, Medical School, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040 Madrid, Spain (E.M.); (S.J.-V.); (J.J.-C.); (C.M.B.); (F.J.C.)
- Hematology Department, IML, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Profesor Martín Lagos s/n, 28040 Madrid, Spain (B.Í.-R.)
| |
Collapse
|
20
|
Antonucci L, Karin M. The Past and Future of Inflammation as a Target to Cancer Prevention. Cancer Prev Res (Phila) 2024; 17:141-155. [PMID: 38271694 PMCID: PMC10987280 DOI: 10.1158/1940-6207.capr-23-0423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/21/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024]
Abstract
Inflammation is an essential defense mechanism in which innate immune cells are coordinately activated on encounter of harmful stimuli, including pathogens, tissue injury, and toxic compounds and metabolites to neutralize and eliminate the instigator and initiate healing and regeneration. Properly terminated inflammation is vital to health, but uncontrolled runaway inflammation that becomes chronic begets a variety of inflammatory and metabolic diseases and increases cancer risk. Making damaged tissues behave as "wounds that do not heal" and sustaining the production of growth factors whose physiologic function is tissue healing, chronic inflammation accelerates cancer emergence from premalignant lesions. In 1863, Rudolf Virchow, a leading German pathologist, suggested a possible association between inflammation and tumor formation, but it took another 140 years to fully elucidate and appreciate the tumorigenic role of inflammation. Key findings outlined molecular events in the inflammatory cascade that promote cancer onset and progression and enabled a better appreciation of when and where inflammation should be inhibited. These efforts triggered ongoing research work to discover and develop inflammation-reducing chemopreventive strategies for decreasing cancer risk and incidence.
Collapse
Affiliation(s)
- Laura Antonucci
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine; La Jolla, CA 92093, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine; La Jolla, CA 92093, USA
| |
Collapse
|
21
|
Rawat SG, Tiwari RK, Kumar A. Blockade of phosphodiesterase 5 by sildenafil reduces tumor growth and potentiates tumor-killing ability of cisplatin in vivo against T cell lymphoma: Implication of modulated apoptosis, reactive oxygen species homeostasis, glucose metabolism, and pH regulation. ENVIRONMENTAL TOXICOLOGY 2024; 39:1909-1922. [PMID: 38059649 DOI: 10.1002/tox.24074] [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: 07/07/2023] [Revised: 08/25/2023] [Accepted: 11/12/2023] [Indexed: 12/08/2023]
Abstract
In the past years, PDE5 has emerged as a promising therapeutic target for many cancers due to its highly upregulated expression. Interestingly, a recent in vitro study by our group has shown the antitumor and chemopotentiating action of sildenafil against T cell lymphoma. Our study showed that lower doses of sildenafil (50 μM) and cisplatin (0.5 μg/mL) exhibited 4% and 23% cytotoxicity against HuT78 cells, respectively, which was dramatically increased up to 50% when treated with both. Hence, the present study was designed to evaluate the antitumor and chemo-potentiating action of sildenafil in a murine model of T cell lymphoma (popularly called as Dalton's lymphoma [DL]). In the present study, DL-bearing mice were administered with vehicle (PBS), sildenafil (5 mg/kg bw), cisplatin (5 mg/kg bw), and sildenafil and cisplatin followed by evaluation of their impact on tumor growth by analyzing various parameters. The apoptosis was assessed by Wright-Giemsa, annexin-V, and DAPI staining. Reactive oxygen species (ROS) level was examined through DCFDA staining. The expression of genes and proteins were estimated by RT-PCR and Western blotting, respectively. The experimental findings of the study demonstrate for the first time that sildenafil inhibits tumor growth and potentiates tumor inhibitory ability of cisplatin by altering apoptosis, glycolysis, ROS homeostasis, and pH regulation in T cell lymphoma-carrying host. In addition, our investigation also showed amelioration of tumor-induced liver and kidney damage by sildenafil. Overall, the experimental data of our study strongly advocate the use and repurposing of SDF in designing promising chemotherapeutic regimens against malignancies of T cells.
Collapse
Affiliation(s)
- Shiv Govind Rawat
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Rajan Kumar Tiwari
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ajay Kumar
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
22
|
Raulo K, Qazi S. Identification of Key Genes Associated with Polycystic Ovarian Syndrome and Endometrial and Ovarian Cancer through Bioinformatics. J Hum Reprod Sci 2024; 17:81-93. [PMID: 39091436 PMCID: PMC11290717 DOI: 10.4103/jhrs.jhrs_44_24] [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: 03/19/2024] [Revised: 04/13/2024] [Accepted: 06/03/2024] [Indexed: 08/04/2024] Open
Abstract
Background Polycystic ovary syndrome (PCOS), a common endocrine disorder, is linked to increased risks of endometrial cancer (EC) and ovarian cancer (OC). Our study utilises bioinformatics analysis to identify shared gene signatures and elucidate biological processes between EC and OC and PCOS. Aim The objective of this research is to unveil the common molecular landscape shared by PCOS and EC and OC. Settings and Design An observational computational bioinformatics analysis. Materials and Methods Gene expression profiles for PCOS (GSE199225), EC (GSE215413) and OC (GSE174670) were obtained from the Gene Expression Omnibus database. Hub genes were identified through functional enrichment analysis and protein-protein interaction. Drug identification analyses were employed to find drugs targeting the hub genes. Results Key hub genes linking PCOS and EC includes RECQL4, RAD54L, ATR, CHTF18, WDHD1, CDT1, PLK1, PKMYT1, RAD18 and RPL3; for PCOS and OC, they include HMOX1, TXNRD1, NQO1, GCLC, GSTP1, PRDX1, SOD1, GPX3, BOP1 and BYSL. Gene Ontology analysis revealed DNA metabolic process in PCOS and EC, while in PCOS and OC, it identified the removal of superoxide radicals. Kyoto Encyclopaedia of Genes and Genomes pathway analysis highlighted cell cycle in PCOS and EC and hepatocellular carcinoma in PCOS and OC. Potential drugs for PCOS and EC include quercetin, calcitriol and testosterone; for PCOS and OC, eugenol and 1-chloro-2,4-dinitrobenzene are identified. Conclusion These findings offer insights into potential therapeutic targets and pathways linking PCOS with EC and OC, enhancing our understanding of the molecular mechanisms involved in these associations.
Collapse
Affiliation(s)
- Karishma Raulo
- Department of Cancer Biology, Quick is Cool Aitele Research LLP, Bihar, India
| | - Sahar Qazi
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
23
|
Singh P, Dhole B, Choudhury J, Tuli A, Pandey D, Velpandian T, Gupta S, Chaturvedi PK. Calotropis procera extract inhibits prostate cancer through regulation of autophagy. J Cell Mol Med 2024; 28:e18050. [PMID: 38400579 PMCID: PMC10941509 DOI: 10.1111/jcmm.18050] [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: 08/29/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 02/25/2024] Open
Abstract
Current treatment options available for prostate cancer (PCa) patients have many adverse side effects and hence, new alternative therapies need to be explored. Anticancer potential of various phytochemicals derived from Calotropis procera has been studied in many cancers but no study has investigated the effect of leaf extract of C. procera on PCa cells. Hence, we investigated the effect of C. procera leaf extract (CPE) on cellular properties of androgen-independent PC-3 and androgen-sensitive 22Rv1 cells. A hydroalcoholic extract of C. procera was prepared and MTT assay was performed to study the effect of CPE on viability of PCa cells. The effect of CPE on cell division ability, migration capability and reactive oxygen species (ROS) production was studied using colony formation assay, wound-healing assay and 2',7'-dichlorodihydrofluorescein diacetate assay, respectively. Caspase activity assay and LDH assay were performed to study the involvement of apoptosis and necrosis in CPE-mediated cell death. Protein levels of cell cycle, antioxidant, autophagy and apoptosis markers were measured by western blot. The composition of CPE was identified using untargeted LC-MS analysis. Results showed that CPE decreased the viability of both the PCa cells, PC-3 and 22Rv1, in a dose- and time-dependent manner. Also, CPE significantly inhibited the colony-forming ability, migration and endogenous ROS production in both the cell lines. Furthermore, CPE significantly decreased NF-κB protein levels and increased the protein levels of the cell cycle inhibitor p27. A significant increase in expression of autophagy markers was observed in CPE-treated PC-3 cells while autophagy markers were downregulated in 22Rv1 cells after CPE exposure. Hence, it can be concluded that CPE inhibits PCa cell viability possibly by regulating the autophagy pathway and/or altering the ROS levels. Thus, CPE can be explored as a possible alternative therapeutic agent for PCa.
Collapse
Affiliation(s)
- Palak Singh
- Department of Reproductive BiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Bodhana Dhole
- Department of Reproductive BiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Jaganmoy Choudhury
- Department of Reproductive BiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Anannya Tuli
- Department of Ocular PharmacologyDr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical SciencesNew DelhiIndia
| | - Deepak Pandey
- Department of Reproductive BiologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Thirumurthy Velpandian
- Department of Ocular PharmacologyDr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical SciencesNew DelhiIndia
| | - Surabhi Gupta
- Department of Reproductive BiologyAll India Institute of Medical SciencesNew DelhiIndia
| | | |
Collapse
|
24
|
Li S, Cao C, Huang Z, Tang D, Chen J, Wang A, He Q. SOD2 confers anlotinib resistance via regulation of mitochondrial damage in OSCC. Oral Dis 2024; 30:281-291. [PMID: 36229195 DOI: 10.1111/odi.14404] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/22/2022] [Accepted: 10/03/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Previous studies had revealed that anlotinib had outstanding anti-tumor efficacy on oral squamous cell carcinoma. However, the underlying mechanism is still unclear. MATERIALS AND METHODS Anlotinib resistant OSCC cells were established and analyzed by RNA-sequencing. The correlations between SOD2 expression and anlotinib resistance were investigated in OSCC cells and PDX models. Functional assays were performed to verify the SOD2 expression and anlotinib resistance in OSCC cells. RESULTS Anlotinib resistant genes were enriched in the biological processes of mitochondrion organization and the gene pathway of reactive oxygen species. SOD2 expression level was positively correlated with the resistance of anlotinib in OSCC cells and PDX models. Higher SOD2 expression of OSCC cells was more resistant to anlotinib. Anlotinib induced ROS generation, apoptosis and mitochondrial damage in OSCC cells, which can be enhanced by SOD2 knockdown and decreased by SOD2 overexpression. Mitochondrial damage was identified as swelling and cristae disappearance morphology under TEM, decreased mitochondrial membrane potential and lower MFN2 expression. CONCLUSIONS SOD2 may be capable of protecting mitochondria by downregulating ROS generation, which contributes to the resistance of anlotinib in OSCC cells. SOD2 can be utilized as a potential therapeutic target to improve the anti-cancer efficacy of anlotinib in OSCC.
Collapse
Affiliation(s)
- Shuai Li
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, China
| | - Congyuan Cao
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhexun Huang
- Center of Oral Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Dongxiao Tang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Chen
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Anxun Wang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qianting He
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
25
|
Dash S, Ueda T, Komuro A, Honda M, Sugisawa R, Okada H. Deoxycytidine kinase inactivation enhances gemcitabine resistance and sensitizes mitochondrial metabolism interference in pancreatic cancer. Cell Death Dis 2024; 15:131. [PMID: 38346958 PMCID: PMC10861559 DOI: 10.1038/s41419-024-06531-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is considered one of the most lethal forms of cancer. Although in the last decade, an increase in 5-year patient survival has been observed, the mortality rate remains high. As a first-line treatment for PDAC, gemcitabine alone or in combination (gemcitabine plus paclitaxel) has been used; however, drug resistance to this regimen is a growing issue. In our previous study, we reported MYC/glutamine dependency as a therapeutic target in gemcitabine-resistant PDAC secondary to deoxycytidine kinase (DCK) inactivation. Moreover, enrichment of oxidative phosphorylation (OXPHOS)-associated genes was a common property shared by PDAC cell lines, and patient clinical samples coupled with low DCK expression was also demonstrated, which implicates DCK in cancer metabolism. In this article, we reveal that the expression of most genes encoding mitochondrial complexes is remarkably upregulated in PDAC patients with low DCK expression. The DCK-knockout (DCK KO) CFPAC-1 PDAC cell line model reiterated this observation. Particularly, OXPHOS was functionally enhanced in DCK KO cells as shown by a higher oxygen consumption rate and mitochondrial ATP production. Electron microscopic observations revealed abnormal mitochondrial morphology in DCK KO cells. Furthermore, DCK inactivation exhibited reactive oxygen species (ROS) reduction accompanied with ROS-scavenging gene activation, such as SOD1 and SOD2. SOD2 inhibition in DCK KO cells clearly induced cell growth suppression. In combination with increased anti-apoptotic gene BCL2 expression in DCK KO cells, we finally reveal that venetoclax and a mitochondrial complex I inhibitor are therapeutically efficacious for DCK-inactivated CFPAC-1 cells in in vitro and xenograft models. Hence, our work provides insight into inhibition of mitochondrial metabolism as a novel therapeutic approach to overcome DCK inactivation-mediated gemcitabine resistance in PDAC patient treatment.
Collapse
Affiliation(s)
- Suman Dash
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Takeshi Ueda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Akiyoshi Komuro
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Masahiko Honda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Ryoichi Sugisawa
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Hitoshi Okada
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan.
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan.
- Anti-aging Center, Kindai University, Higashi-Osaka, Osaka, 577-8502, Japan.
| |
Collapse
|
26
|
Liang X, Wang Z, Dai Z, Liu J, Zhang H, Wen J, Zhang N, Zhang J, Luo P, Liu Z, Liu Z, Cheng Q. Oxidative stress is involved in immunosuppression and macrophage regulation in glioblastoma. Clin Immunol 2024; 258:109802. [PMID: 37866784 DOI: 10.1016/j.clim.2023.109802] [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: 04/25/2022] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023]
Abstract
Oxidative stress dually affected cancer progression, while its effect on glioblastomas remained unclear. Herein, we clustered the multicenter glioblastoma cohorts based on the oxidative-stress-responsive genes (OSS) expression. We found that cluster 2 with high OSS levels suffered a worse prognosis. Functional analyses and immune-related analyses results exhibited that M2-like pro-tumoral macrophages and neutrophils were enriched in cluster 2, while Natural killer cells' infiltration was decreased. The increased M2-like pro-tumoral macrophages in cluster 2 was confirmed by immunofluorescence. An integrated single-cell analysis validated the malignant features of cluster 2 neoplastic cells and discovered their crosstalk with M2-like pro-tumoral macrophages. Moreover, we observed that SOD3 knockdown might decrease the M2-like pro-tumoral transformation of macrophage in vitro and in vivo. Comprehensively, we revealed oxidative stress' prognostic and immunosuppressive potential in glioblastoma and discovered SOD3's potential role in regulating macrophage M2-like pro-tumoral transformation.
Collapse
Affiliation(s)
- Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China
| | - Jian Liu
- Experiment Center of Medical Innovation, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China
| | - Jie Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, Guangdong, PR China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, Guangdong, PR China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, PR China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China; Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, PR China.
| |
Collapse
|
27
|
Dos Santos AV, Kaul AJ, Dos Santos GT, Dal Berto M, Manfroi LM, Rizzotto G, Roehe AV, Alves RCS, Lutz A, Beck P, Alves RJV, Cruz IBM, Bica CG. The impact of the association between Val16Ala-SOD2 SNP and SOD2 immunohistochemistry expression in the prognosis of patients with esophageal cancer. Pathol Res Pract 2024; 253:154965. [PMID: 38039740 DOI: 10.1016/j.prp.2023.154965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 12/03/2023]
Abstract
INTRODUCTION Esophageal cancer is an extensive public health issue worldwide, warranting the search for biomarkers related to its risk and progression. Previous studies have indicated an association between Val16AlaSOD2 single nucleotide polymorphism in the gene encoding the enzyme superoxide dismutase 2 and esophageal cancer. However, further investigations are needed to clarify its role in disease risk and progression. OBJECTIVE To investigate the role of Val16AlaSOD2-SNP in esophageal cancer progression and in the survival of patients METHODS: Tumor samples were utilized for Val16Ala-SNP genotyping, while SOD2 expression levels in tissue were assessed using immunohistochemistry. A SOD2 Val16Ala-SNP database was used to obtain information on the genotype of healthy individuals. Risk and overall survival analyzes were performed. RESULTS The Val16Ala SNP was associated with an increased risk of esophageal cancer (RR 2.18, 95%CI 1.23-3.86), regardless of age and gender, but did not have a significant effect on patient survival. In contrast, weak SOD2 expression demonstrated a significantly associated with poor overall survival after treatment, independent of other clinicopathological variables (HR, 0.41; 95% CI, 0.22-0.79 P = 0.007). CONCLUSIONS Val16Ala SNP was positively associated with esophageal cancer, and the expression of SOD2 was an independent prognostic marker.
Collapse
Affiliation(s)
- A V Dos Santos
- Graduate Program in Pathology, Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil.
| | - A J Kaul
- Biomedice School, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - G T Dos Santos
- Graduate Program in Pathology, Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - M Dal Berto
- Graduate Program in Pathology, Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - L M Manfroi
- Medical School, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - G Rizzotto
- Laboratory of Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - A V Roehe
- Graduate Program in Pathology, Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - R C S Alves
- Graduate Program in Pathology, Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - A Lutz
- Clinical Oncology Department, Hospital Santa Rita, Complexo Hospitalar Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - P Beck
- Clinical Oncology Department, Hospital Santa Rita, Complexo Hospitalar Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - R J V Alves
- Clinical Oncology Department, Hospital Santa Rita, Complexo Hospitalar Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Department of Internal Medicine, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - I B M Cruz
- Biogenomics Laboratory, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - C G Bica
- Graduate Program in Pathology, Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
28
|
Atiya A, Muhsinah AB, Alrouji M, Alhumaydhi FA, Al Abdulmonem W, Aljasir MA, Sharaf SE, Furkan M, Khan RH, Shahwan M, Shamsi A. Unveiling promising inhibitors of superoxide dismutase 1 (SOD1) for therapeutic interventions. Int J Biol Macromol 2023; 253:126684. [PMID: 37666395 DOI: 10.1016/j.ijbiomac.2023.126684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Superoxide dismutase 1 (SOD1) is a vital enzyme responsible for controlling cellular oxidative stress. Any dysregulation of SOD1 activity is linked with cancer pathogenesis and neurodegenerative disorders, such as amyotrophic lateral sclerosis (ALS). Among the inhibitors known to be effective against SOD1, LCS-1 stands out; however, its efficacy, specificity, and safety profiles are somewhat restricted. In this study, we used PubChem library to retrieve compounds that exhibited a structural similarity of at least 90 % with LCS-1. These compounds underwent molecular docking analyses to examine their interaction patterns and binding affinities with SOD1. Further, we applied filters based on physicochemical and ADMET properties, refining the selection process. Our analysis revealed that selected compounds interact with crucial residues of SOD1 active site. To gain further insights into conformational stability and dynamics of the SOD1-ligand complexes, we conducted all-atom molecular dynamics (MD) simulations for 100 ns. We identified two compounds, CID:133306073 and CID:133446715, as potential scaffolds with promising inhibitory properties against SOD1. Both compounds hold significant potential for further exploration as therapeutic SOD1 inhibitors. Further studies are warranted to fully harness their therapeutic potential in targeting SOD1 for cancer and ALS treatment, offering new avenues for improved patient outcomes and disease management.
Collapse
Affiliation(s)
- Akhtar Atiya
- Department of Pharmacognosy, College of Pharmacy, King Khalid University (KKU), Guraiger St., Abha 62529, Saudi Arabia.
| | - Abdullatif Bin Muhsinah
- Department of Pharmacognosy, College of Pharmacy, King Khalid University (KKU), Guraiger St., Abha 62529, Saudi Arabia; Complementary and Alternative Medicine Unit, King Khalid University (KKU), Guraiger St., Abha 62529, Saudi Arabia.
| | - Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia.
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia.
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraydah 51452, Saudi Arabia.
| | - Mohammad A Aljasir
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia.
| | - Sharaf E Sharaf
- Pharmaceutical Chemistry Department, College of Pharmacy Umm Al-Qura University Makkah, Saudi Arabia.
| | - Mohammad Furkan
- Department of Biochemistry, Aligarh Muslim University, Aligarh, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Moyad Shahwan
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, P.O. Box 346, United Arab Emirates
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, P.O. Box 346, United Arab Emirates.
| |
Collapse
|
29
|
Arnhold J. Inflammation-Associated Cytotoxic Agents in Tumorigenesis. Cancers (Basel) 2023; 16:81. [PMID: 38201509 PMCID: PMC10778456 DOI: 10.3390/cancers16010081] [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: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic inflammatory processes are related to all stages of tumorigenesis. As inflammation is closely associated with the activation and release of different cytotoxic agents, the interplay between cytotoxic agents and antagonizing principles is highlighted in this review to address the question of how tumor cells overcome the enhanced values of cytotoxic agents in tumors. In tumor cells, the enhanced formation of mitochondrial-derived reactive species and elevated values of iron ions and free heme are antagonized by an overexpression of enzymes and proteins, contributing to the antioxidative defense and maintenance of redox homeostasis. Through these mechanisms, tumor cells can even survive additional stress caused by radio- and chemotherapy. Through the secretion of active agents from tumor cells, immune cells are suppressed in the tumor microenvironment and an enhanced formation of extracellular matrix components is induced. Different oxidant- and protease-based cytotoxic agents are involved in tumor-mediated immunosuppression, tumor growth, tumor cell invasion, and metastasis. Considering the special metabolic conditions in tumors, the main focus here was directed on the disturbed balance between the cytotoxic agents and protective mechanisms in late-stage tumors. This knowledge is mandatory for the implementation of novel anti-cancerous therapeutic approaches.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
30
|
Wang Q, Zhong Y, Li N, Du L, Ye R, Xie Y, Hu F. Combination of dimethylmethoxy chromanol and turmeric root extract synergically attenuates ultraviolet-induced oxidative damage by increasing endogenous antioxidants in HaCaT cells. Skin Res Technol 2023; 29:e13539. [PMID: 38115632 PMCID: PMC10730978 DOI: 10.1111/srt.13539] [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: 10/01/2023] [Accepted: 11/16/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Repeated exposure to UV generates excessive reactive oxygen species (ROS) and damages the enzymatic antioxidant defense system including quinone oxidoreductase 1 (NQO1) and superoxide dismutase (SOD) in skin. Topical application of antioxidants may prevent the undesired damage of cellular proteins, lipids and DNA in skin. Dimethylmethoxy chromanol (DMC) is a bioinspired molecule, designed to be a structural analog to the γ-tocopherol that is naturally present in vegetables and plants. Turmeric root extract (TRE) is from a plant in South Asia extensively used as a food spice & vegetable, and its main components are turmerones. As both DMC and TRE are strong antioxidants with complementary antioxidation mechanisms, the aim of this study was to investigate the enhanced protective effects of their combination on oxidative damage in HaCaT cells following UVB exposure. MATERIALS AND METHODS The effects of single and combined administrations of DMC and TRE on the SOD activity of HaCaT cells were evaluated by the SOD assay and qPCR. The NQO1 expression in the UVB-treated HaCaT cells was analyzed by the Western Blot. Furthermore, a clinical test involving 24 subjects was conducted to evaluate the in vivo antioxidation efficacies of the serum formulated with the combination of DMC and TRE at the optimal weight ratio. RESULTS SOD assay showed that pretreating DMC or TRE alone could not preserve the impaired HaCaT SOD activity after UVB treatment. DMC and TRE at 1:1 weight ratio was the optimal combination to enhance the HaCaT SOD activity by approximately more than 1-fold compared with either of the single treated groups. No enhancement effect was observed at other mixing ratios. The 1:1 weight ratio was further proved to be optimal as this combination boosted the NQO1 expression by more than 50%, whereas no boosting effect was observed at other mixing ratios. The clinical test of the serum containing this optimal antioxidant combination demonstrated promising in vivo antioxidation efficacies after 4-week use, including 7.16% improvement in skin lightening, 18.29% reduction in skin redness, 35.68% decrease in TEWL, 19.05% increase in skin gloss and 32.04% enhancement in skin firmness. CONCLUSION Collectively, our results indicated that the combination of DMC and TRE at 1:1 weight ratio attenuated the UV-induced oxidative damage by synergistically boosting endogenous antioxidant enzyme activity in HaCaT cells. Therefore, this optimal antioxidant combination is a promising treatment to boost skin antioxidation defense system.
Collapse
Affiliation(s)
- Qianqian Wang
- Department of DermatologyHuashan HospitalFudan UniversityShanghaiChina
| | - Ye Zhong
- UNISKIN Research Institute on Skin AgingInertia Shanghai Biotechnology Co., Ltd.ShanghaiChina
- DermaHealth Shanghai Biotechnology Co., Ltd.ShanghaiChina
| | - Nihong Li
- UNISKIN Research Institute on Skin AgingInertia Shanghai Biotechnology Co., Ltd.ShanghaiChina
- DermaHealth Shanghai Biotechnology Co., Ltd.ShanghaiChina
| | - Le Du
- UNISKIN Research Institute on Skin AgingInertia Shanghai Biotechnology Co., Ltd.ShanghaiChina
- DermaHealth Shanghai Biotechnology Co., Ltd.ShanghaiChina
| | - Rui Ye
- UNISKIN Research Institute on Skin AgingInertia Shanghai Biotechnology Co., Ltd.ShanghaiChina
- DermaHealth Shanghai Biotechnology Co., Ltd.ShanghaiChina
| | - Yicheng Xie
- Department of DermatologyThe Children's HospitalZhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang UniversityHangzhouChina
| | - Fan Hu
- UNISKIN Research Institute on Skin AgingInertia Shanghai Biotechnology Co., Ltd.ShanghaiChina
- DermaHealth Shanghai Biotechnology Co., Ltd.ShanghaiChina
| |
Collapse
|
31
|
Lazzarini R, Tartaglione MF, Ciarapica V, Piva F, Giulietti M, Fulgenzi G, Martelli M, Ledda C, Vitale E, Malavolta M, Santarelli L, Bracci M. Keratinocytes Exposed to Blue or Red Light: Proteomic Characterization Showed Cytoplasmic Thioredoxin Reductase 1 and Aldo-Keto Reductase Family 1 Member C3 Triggered Expression. Int J Mol Sci 2023; 24:16189. [PMID: 38003379 PMCID: PMC10671521 DOI: 10.3390/ijms242216189] [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: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Several cell-signaling mechanisms are activated by visible light radiation in human keratinocytes, but the key regulatory proteins involved in this specific cellular response have not yet been identified. Human keratinocytes (HaCaT cells) were exposed to blue or red light at low or high irradiance for 3 days in cycles of 12 h of light and 12 h of dark. The cell viability, apoptotic rate and cell cycle progression were analyzed in all experimental conditions. The proteomic profile, oxidative stress and mitochondrial morphology were additionally evaluated in the HaCaT cells following exposure to high-irradiance blue or red light. Low-irradiance blue or red light exposure did not show an alteration in the cell viability, cell death or cell cycle progression. High-irradiance blue or red light reduced the cell viability, induced cell death and cell cycle G2/M arrest, increased the reactive oxygen species (ROS) and altered the mitochondrial density and morphology. The proteomic profile revealed a pivotal role of Cytoplasmic thioredoxin reductase 1 (TXNRD1) and Aldo-keto reductase family 1 member C3 (AKR1C3) in the response of the HaCaT cells to high-irradiance blue or red light exposure. Blue or red light exposure affected the viability of keratinocytes, activating a specific oxidative stress response and inducing mitochondrial dysfunction. Our results can help to address the targets for the therapeutic use of light and to develop adequate preventive strategies for skin damage. This in vitro study supports further in vivo investigations of the biological effects of light on human keratinocytes.
Collapse
Affiliation(s)
- Raffaella Lazzarini
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy; (M.F.T.); (V.C.); (M.M.); (M.B.)
| | - Maria Fiorella Tartaglione
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy; (M.F.T.); (V.C.); (M.M.); (M.B.)
| | - Veronica Ciarapica
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy; (M.F.T.); (V.C.); (M.M.); (M.B.)
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (F.P.)
| | - Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (F.P.)
| | - Gianluca Fulgenzi
- Department of Clinical and Molecular Sciences Experimental Pathology, Polytechnic University of Marche, 60126 Ancona, Italy;
| | - Margherita Martelli
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy; (M.F.T.); (V.C.); (M.M.); (M.B.)
| | - Caterina Ledda
- Section of Occupational Medicine, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy;
| | - Ermanno Vitale
- Faculty of Medicine and Surgery, Kore University, 94100 Enna, Italy;
| | - Marco Malavolta
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, Scientific Technological Area, IRCCS INRCA, 60121 Ancona, Italy;
| | - Lory Santarelli
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy; (M.F.T.); (V.C.); (M.M.); (M.B.)
| | - Massimo Bracci
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy; (M.F.T.); (V.C.); (M.M.); (M.B.)
| |
Collapse
|
32
|
Lee C, Kim HN, Kwon JA, Hwang J, Park JY, Shin OS, Yoon SY, Yoon J. Identification of a Complex Karyotype Signature with Clinical Implications in AML and MDS-EB Using Gene Expression Profiling. Cancers (Basel) 2023; 15:5289. [PMID: 37958462 PMCID: PMC10648390 DOI: 10.3390/cancers15215289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Complex karyotype (CK) is associated with a poor prognosis in both acute myeloid leukemia (AML) and myelodysplastic syndrome with excess blasts (MDS-EB). Transcriptomic analyses have improved our understanding of the disease and risk stratification of myeloid neoplasms; however, CK-specific gene expression signatures have been rarely investigated. In this study, we developed and validated a CK-specific gene expression signature. Differential gene expression analysis between the CK and non-CK groups using data from 348 patients with AML and MDS-EB from four cohorts revealed enrichment of the downregulated genes localized on chromosome 5q or 7q, suggesting that haploinsufficiency due to the deletion of these chromosomes possibly underlies CK pathogenesis. We built a robust transcriptional model for CK prediction using LASSO regression for gene subset selection and validated it using the leave-one-out cross-validation method for fitting the logistic regression model. We established a 10-gene CK signature (CKS) predictive of CK with high predictive accuracy (accuracy 94.22%; AUC 0.977). CKS was significantly associated with shorter overall survival in three independent cohorts, and was comparable to that of previously established risk stratification models for AML. Furthermore, we explored of therapeutic targets among the genes comprising CKS and identified the dysregulated expression of superoxide dismutase 1 (SOD1) gene, which is potentially amenable to SOD1 inhibitors.
Collapse
Affiliation(s)
- Cheonghwa Lee
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul 08308, Republic of Korea; (C.L.); (H.N.K.); (J.A.K.); (J.H.)
| | - Ha Nui Kim
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul 08308, Republic of Korea; (C.L.); (H.N.K.); (J.A.K.); (J.H.)
| | - Jung Ah Kwon
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul 08308, Republic of Korea; (C.L.); (H.N.K.); (J.A.K.); (J.H.)
| | - Jinha Hwang
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul 08308, Republic of Korea; (C.L.); (H.N.K.); (J.A.K.); (J.H.)
| | - Ji-Ye Park
- BK21 Graduate Program, Department of Biomedical Sciences, College of Medicine, Korea University Guro Hospital, Seoul 08308, Republic of Korea (O.S.S.)
| | - Ok Sarah Shin
- BK21 Graduate Program, Department of Biomedical Sciences, College of Medicine, Korea University Guro Hospital, Seoul 08308, Republic of Korea (O.S.S.)
| | - Soo-Young Yoon
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul 08308, Republic of Korea; (C.L.); (H.N.K.); (J.A.K.); (J.H.)
| | - Jung Yoon
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul 08308, Republic of Korea; (C.L.); (H.N.K.); (J.A.K.); (J.H.)
| |
Collapse
|
33
|
Lei G, Tang L, Yu Y, Bian W, Yu L, Zhou J, Li Y, Wang Y, Du J. The potential of targeting cuproptosis in the treatment of kidney renal clear cell carcinoma. Biomed Pharmacother 2023; 167:115522. [PMID: 37757497 DOI: 10.1016/j.biopha.2023.115522] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/07/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Renal cell carcinoma (RCC) is one of the top ten malignancies and tumor-related causes of death worldwide. The most common histologic subtype is kidney renal clear cell carcinoma (KIRC), accounting for approximately 75% of all RCC cases. Early resection is considered the basic treatment for patients with KIRC. However, approximately 30% of these patients experience recurrence post-operation. Cuproptosis, an autonomous mechanism for controlling cell death, encompasses various molecular mechanisms and multiple cellular metabolic pathways. These pathways mainly include copper metabolic signaling pathways, mitochondrial metabolism signaling pathways, and lipoic acid pathway signaling pathways. Recent evidence shows that cuproptosis is identified as a key cell death modality that plays a meaningful role in tumor progression. However, there is no published systematic review that summarizes the correlation between cuproptosis and KIRC, despite the fact that investigations on cuproptosis and the pathogenesis of KIRC have increased in past years. Researchers have discovered that exogenous copper infusion accelerates the dysfunction of mitochondrial dysfunction and suppresses KIRC cells by inducing cuproptosis. The levels of tricarboxylic acid cycle proteins, lipoic acid protein, copper, and ferredoxin 1 (FDX1) were dysregulated in KIRC cells, and the prognosis of patients with high FDX1 expression is better than that of patients with low expression. Cuproptosis played an indispensable role in the regulation of tumor microenvironment features, tumor progression, and long-term prognosis of KIRC. In this review, we summarized the systemic and cellular metabolic processes of copper and the copper-related signaling pathways, highlighting the potential targets related to cuproptosis for KIRC treatment.
Collapse
Affiliation(s)
- Guojie Lei
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Lusheng Tang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yanhua Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wenxia Bian
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Lingyan Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Junyu Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China.
| | - Ying Wang
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
34
|
Radwan SM, Alqulaly M, Elsaeed MY, Elshora SZ, Atwa AH, Wasfey EF. L-carnitine reverses methotrexate-induced nephrotoxicity in experimental rat model: Insight on SIRT1/PGC-1α/Nrf2/HO-1 axis. J Appl Toxicol 2023; 43:1667-1675. [PMID: 37312617 DOI: 10.1002/jat.4503] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023]
Abstract
Methotrexate (MTX) is a chemotherapeutic agent used for treating several types of cancer as well as psoriasis and rheumatoid arthritis, but its use is limited due to its nephrotoxicity. The purpose of this research work was to observe ameliorative effects of L-carnitine (LC) toward renal toxicity caused by MTX and mechanisms responsible for these effects. Thirty-two male Sprague-Dawley rats were divided into four groups (eight rats/group), control group (received saline), MTX group (20 mg/kg/i.p. once), LC group (500 mg/kg/i.p. for 5 days), and MTX + LC group (received a single MTX dose 20 mg/kg/i.p. followed by LC 500 mg/kg/i.p. for 5 days). Histopathological examinations, lipid oxidation marker, malondialdehyde (MDA), and the antioxidant superoxide dismutase (SOD) as well as inflammatory (tumor necrosis factor-α [TNF-α] and interleukin-6 [IL-6]) and apoptotic markers (Bax, Bcl2, and caspase-3) were used to assess renal toxicity. Moreover, the protein levels of silent information regulator 1 (SIRT1) and its downstream signaling targets, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), and nuclear factor erythroid 2-related factor 2 (Nrf2) in addition to heme oxygenase-1 (HO-1) were measured. LC significantly protected against MTX-induced nephrotoxicity. It ameliorated MTX-induced renal histopathological changes and diminished MTX-induced renal oxidative stress, renal inflammation, and apoptosis. LC also upregulated the expression of SIRT1 and PGC-1 as well as Nrf2 and HO-1. By controlling the expression of renal SIRT1/PGC-1/Nrf2/HO-1, LC displayed antioxidant, anti-inflammatory, and anti-apoptotic activities. Hence, using LC supplements may help prevent negative MTX side effects.
Collapse
Affiliation(s)
- Sara M Radwan
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mustafa Alqulaly
- Physiology Department, Damietta Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Magdy Y Elsaeed
- Physiology Department, Damietta Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Shimaa Z Elshora
- Histology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Asmaa H Atwa
- Forensic medicine and clinical toxicology Department, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Eman F Wasfey
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
35
|
Alcolea Palafox M, Belskaya NP, Todorov LT, Kostova IP. Structural Study of a La(III) Complex of a 1,2,3-Triazole Ligand with Antioxidant Activity. Antioxidants (Basel) 2023; 12:1872. [PMID: 37891952 PMCID: PMC10604163 DOI: 10.3390/antiox12101872] [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: 09/20/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
The 1,2,3-triazole derivative 2-(4-chlorophenyl)-5-(pyrrolidin-1-yl)-2H-1,2,3-triazole-4-carboxylic acid with potential anticancer activity was used as a ligand in complex formation with the lanthanum(III) ion. The molecular structure and vibrational spectra of the complex were optimized at three DFT levels, and the scaled IR and Raman spectra were compared to the experimental ones. Several scaling procedures were used. Through a detailed analysis, the structure predicted for the newly synthetized La(III) complex was confirmed by the good accordance of the calculated/experimental IR and Raman spectra. The best DFT method appeared to be M06-2X with the Lanl2mb basis set, followed closely by Lanl2dz. The effect of the lanthanide atom on the molecular structure and atomic charge distribution of the triazole ring was evaluated. The potential free radical scavenging activity of both the ligand and the complex was investigated in several radical-generating model systems. The potential mechanisms of antioxidant action (hydrogen atom transfer (HAT) and single-electron transfer (SET)) were elucidated.
Collapse
Affiliation(s)
- Mauricio Alcolea Palafox
- Department of Physical Chemistry, Faculty of Chemical Sciences, Complutense University, 28040 Madrid, Spain
| | - Nataliya P. Belskaya
- Department of Technology for Organic Synthesis, Ural Federal University, 19 Mira Str., Yekaterinburg 620012, Russia;
| | - Lozan T. Todorov
- Department of Chemistry, Faculty of Pharmacy, Medical University—Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria;
| | - Irena P. Kostova
- Department of Chemistry, Faculty of Pharmacy, Medical University—Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria;
| |
Collapse
|
36
|
Jelic M, Mandic A, Maricic S, Bozin B, Kladar N, Sudji J, Conic BS. PREDICTIVE POWER OF OXIDATIVE STRESS BIOMARKERS IN RECURRENCE AND SURVIVAL IN ADVANCED CERVICAL CANCER. Exp Oncol 2023; 45:231-241. [PMID: 37824768 DOI: 10.15407/exp-oncology.2023.02.231] [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: 10/11/2023] [Indexed: 10/14/2023]
Abstract
The aim of our study was to measure the levels of 8-hydroxy-2-deoxyguanosine, malondialdehyde, and antioxidant enzymes in patients with locally advanced cervical cancer prior to treatment to determine how these evaluated biomarkers are associated with cervical cancer recurrence and to estimate their potential in further research and clinical use. MATERIALS AND METHODS The study included 45 female patients with newly diagnosed advanced cervical cancer who underwent concomitant chemoradiotherapy. The blood and urine samples were collected prior to treatment, between December 2013 and April 2016, and subsequent laboratory analysis was performed. After the medium follow-up of 29 months, the patients were divided into 3 groups according to the time of disease recurrence. A statistical analysis was performed in order to evaluate the relationship between the previously measured biomarkers and recurrence. RESULTS Taken individually, the parameters of oxidative stress did not reveal significant differences between the three groups in our study. Nevertheless, the catalase and glutathione S-transferase activities were the best predictors of the recurrence. Based on the activities of these two oxidative enzymes, it was possible to separate the group of patients without recurrence after follow-up from the other two groups of patients with recurrent disease. CONCLUSIONS The parameters of oxidative stress have a certain predictive value on the outcome of patients with advanced cervical cancer after concomitant chemo-radiotherapy.
Collapse
Affiliation(s)
- Marija Jelic
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Aljosa Mandic
- Oncology Institute of Vojvodina, Sremska Kamenica, Serbia
- University of Novi Sad, Faculty of Medicine, Department of Gynecology and Obstetrics, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Slobodan Maricic
- Oncology Institute of Vojvodina, Sremska Kamenica, Serbia
- University of Novi Sad, Faculty of Medicine, Department of Gynecology and Obstetrics, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Biljana Bozin
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Nebojsa Kladar
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
- University of Novi Sad, Faculty of Medicine, Center for Medical and Pharmaceutical Investigations and Quality Control, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Jan Sudji
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Branislava Srdjenovic Conic
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
- University of Novi Sad, Faculty of Medicine, Center for Medical and Pharmaceutical Investigations and Quality Control, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| |
Collapse
|
37
|
Nizami ZN, Aburawi HE, Semlali A, Muhammad K, Iratni R. Oxidative Stress Inducers in Cancer Therapy: Preclinical and Clinical Evidence. Antioxidants (Basel) 2023; 12:1159. [PMID: 37371889 DOI: 10.3390/antiox12061159] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Reactive oxygen species (ROS) are metabolic byproducts that regulate various cellular processes. However, at high levels, ROS induce oxidative stress, which in turn can trigger cell death. Cancer cells alter the redox homeostasis to facilitate protumorigenic processes; however, this leaves them vulnerable to further increases in ROS levels. This paradox has been exploited as a cancer therapeutic strategy with the use of pro-oxidative drugs. Many chemotherapeutic drugs presently in clinical use, such as cisplatin and doxorubicin, induce ROS as one of their mechanisms of action. Further, various drugs, including phytochemicals and small molecules, that are presently being investigated in preclinical and clinical studies attribute their anticancer activity to ROS induction. Consistently, this review aims to highlight selected pro-oxidative drugs whose anticancer potential has been characterized with specific focus on phytochemicals, mechanisms of ROS induction, and anticancer effects downstream of ROS induction.
Collapse
Affiliation(s)
- Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| | - Hanan E Aburawi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| | - Abdelhabib Semlali
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire-Université Laval, Quebec, QC G1V 0A6, Canada
| | - Khalid Muhammad
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| |
Collapse
|
38
|
Li C, Wu C, Ji C, Xu G, Chen J, Zhang J, Hong H, Liu Y, Cui Z. The pathogenesis of DLD-mediated cuproptosis induced spinal cord injury and its regulation on immune microenvironment. Front Cell Neurosci 2023; 17:1132015. [PMID: 37228705 PMCID: PMC10203164 DOI: 10.3389/fncel.2023.1132015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 04/18/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Spinal cord injury (SCI) is a severe central nervous system injury that leads to significant sensory and motor impairment. Copper, an essential trace element in the human body, plays a vital role in various biological functions and is strictly regulated by copper chaperones and transporters. Cuproptosis, a novel type of metal ion-induced cell death, is distinct from iron deprivation. Copper deprivation is closely associated with mitochondrial metabolism and mediated by protein fatty acid acylation. Methods In this study, we investigated the effects of cuproptosis-related genes (CRGs) on disease progression and the immune microenvironment in acute spinal cord injury (ASCI) patients. We obtained the gene expression profiles of peripheral blood leukocytes from ASCI patients using the Gene Expression Omnibus (GEO) database. We performed differential gene analysis, constructed protein-protein interaction networks, conducted weighted gene co-expression network analysis (WGCNA), and built a risk model. Results Our analysis revealed that dihydrolipoamide dehydrogenase (DLD), a regulator of copper toxicity, was significantly associated with ASCI, and DLD expression was significantly upregulated after ASCI. Furthermore, gene ontology (GO) enrichment analysis and gene set variation analysis (GSVA) showed abnormal activation of metabolism-related processes. Immune infiltration analysis indicated a significant decrease in T cell numbers in ASCI patients, while M2 macrophage numbers were significantly increased and positively correlated with DLD expression. Discussion In summary, our study demonstrated that DLD affects the ASCI immune microenvironment by promoting copper toxicity, leading to increased peripheral M2 macrophage polarization and systemic immunosuppression. Thus, DLD has potential as a promising biomarker for ASCI, providing a foundation for future clinical interventions.
Collapse
Affiliation(s)
- Chaochen Li
- The Affiliated Hospital 2 of Nantong University, Nantong University, The First People’s Hospital of Nantong, Nantong, China
- Key Laboratory for Restoration Mechanism and Clinical Translation of Spinal Cord Injury, Nantong, China
- Research Institute for Spine and Spinal Cord Disease of Nantong University, Nantong, China
| | - Chunshuai Wu
- The Affiliated Hospital 2 of Nantong University, Nantong University, The First People’s Hospital of Nantong, Nantong, China
- Key Laboratory for Restoration Mechanism and Clinical Translation of Spinal Cord Injury, Nantong, China
- Research Institute for Spine and Spinal Cord Disease of Nantong University, Nantong, China
| | - Chunyan Ji
- The Affiliated Hospital 2 of Nantong University, Nantong University, The First People’s Hospital of Nantong, Nantong, China
- Key Laboratory for Restoration Mechanism and Clinical Translation of Spinal Cord Injury, Nantong, China
- Research Institute for Spine and Spinal Cord Disease of Nantong University, Nantong, China
| | - Guanhua Xu
- The Affiliated Hospital 2 of Nantong University, Nantong University, The First People’s Hospital of Nantong, Nantong, China
| | - Jiajia Chen
- The Affiliated Hospital 2 of Nantong University, Nantong University, The First People’s Hospital of Nantong, Nantong, China
| | - Jinlong Zhang
- The Affiliated Hospital 2 of Nantong University, Nantong University, The First People’s Hospital of Nantong, Nantong, China
| | - Hongxiang Hong
- The Affiliated Hospital 2 of Nantong University, Nantong University, The First People’s Hospital of Nantong, Nantong, China
| | - Yang Liu
- The Affiliated Hospital 2 of Nantong University, Nantong University, The First People’s Hospital of Nantong, Nantong, China
| | - Zhiming Cui
- The Affiliated Hospital 2 of Nantong University, Nantong University, The First People’s Hospital of Nantong, Nantong, China
- Key Laboratory for Restoration Mechanism and Clinical Translation of Spinal Cord Injury, Nantong, China
- Research Institute for Spine and Spinal Cord Disease of Nantong University, Nantong, China
| |
Collapse
|
39
|
Angeletti A, Bruschi M, Kajana X, Spinelli S, Verrina E, Lugani F, Caridi G, Murtas C, Candiano G, Prunotto M, Ghiggeri GM. Mechanisms Limiting Renal Tissue Protection and Repair in Glomerulonephritis. Int J Mol Sci 2023; 24:ijms24098318. [PMID: 37176025 PMCID: PMC10179029 DOI: 10.3390/ijms24098318] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Glomerulonephritis are renal disorders resulting from different pathogenic mechanisms (i.e., autoimmunity, complement, inflammatory activation, etc.). Clarifying details of the pathogenic cascade is basic to limit the progression from starting inflammation to degenerative stages. The balance between tissue injury, activation of protective systems and renal tissue repair determines the final outcome. Induction of an oxidative stress is part of glomerular inflammation and activation of protective antioxidant systems has a crucial role in reducing tissue effects. The generation of highly reactive oxygen species can be evaluated in vivo by tracing the inner-layer content of phosphatidyl ethanolamine and phosphatidyl serine in cell membranes. Albumin is the major antioxidant in serum and the level of oxidized albumin is another indirect sign of oxidative stress. Studies performed in Gn, specifically in FSGS, showed a high degree of oxidation in most contexts. High levels of circulating anti-SOD2 antibodies, limiting the detoxyfing activity of SOD2, have been detected in autoimmune Gn(lupus nephritis and membranous nephropathy) in association with persistence of proteinuria and worsening of renal function. In renal transplant, high levels of circulating anti-Glutathione S-transferase antibodies have been correlated with chronic antibody rejection and progressive loss of renal function. Annexins, mainly ANXA1 and ANXA2, play a general anti-inflammatory effect by inhibiting neutrophil functions. Cytosolic ANXA1 is decreased in apoptotic neutrophils of patients with glomerular polyangitis in association with delayed apoptosis that is considered the mechanism for polyangitis. High circulating levels of anti-ANXA1 and anti-ANXA2 antibodies characterize lupus nephritis implying a reduced anti-inflammatory effect. High circulating levels of antibodies targeting Macrophages (anti-FMNL1) have been detected in Gn in association with proteinuria. They potentially modify the intra-glomerular presence of protective macrophages (M2a, M2c) thus acting on the composition of renal infiltrate and on tissue repair.
Collapse
Affiliation(s)
- Andrea Angeletti
- Nephrology, Dialysis and Transplantation Unit, IRCCS, Istituto GianninaGaslini, 16147 Genova, Italy
| | - Maurizio Bruschi
- Nephrology, Dialysis and Transplantation Unit, IRCCS, Istituto GianninaGaslini, 16147 Genova, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, 16126 Genoa, Italy
| | - Xuliana Kajana
- Nephrology, Dialysis and Transplantation Unit, IRCCS, Istituto GianninaGaslini, 16147 Genova, Italy
| | - Sonia Spinelli
- Nephrology, Dialysis and Transplantation Unit, IRCCS, Istituto GianninaGaslini, 16147 Genova, Italy
| | - Enrico Verrina
- Nephrology, Dialysis and Transplantation Unit, IRCCS, Istituto GianninaGaslini, 16147 Genova, Italy
| | - Francesca Lugani
- Nephrology, Dialysis and Transplantation Unit, IRCCS, Istituto GianninaGaslini, 16147 Genova, Italy
| | - Gialuca Caridi
- Nephrology, Dialysis and Transplantation Unit, IRCCS, Istituto GianninaGaslini, 16147 Genova, Italy
| | - Corrado Murtas
- Nephrology and Dialysis Unit, Ospedale Belcolle, 01100 Viterbo, Italy
| | - Giovanni Candiano
- Nephrology, Dialysis and Transplantation Unit, IRCCS, Istituto GianninaGaslini, 16147 Genova, Italy
| | - Marco Prunotto
- Institute of Pharmaceutical Sciences of Western Switzerland, School of Pharmaceutical Sciences, University of Geneva, 1205 Geneva, Switzerland
| | - Gian Marco Ghiggeri
- Nephrology, Dialysis and Transplantation Unit, IRCCS, Istituto GianninaGaslini, 16147 Genova, Italy
| |
Collapse
|
40
|
Zhang S, Wang L, Kang Y, Wu J, Zhang Z. Nanomaterial-based Reactive Oxygen Species Scavengers for Osteoarthritis Therapy. Acta Biomater 2023; 162:1-19. [PMID: 36967052 DOI: 10.1016/j.actbio.2023.03.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/17/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
Reactive oxygen species (ROS) play distinct but important roles in physiological and pathophysiological processes. Recent studies on osteoarthritis (OA) have suggested that ROS plays a crucial role in its development and progression, serving as key mediators in the degradation of the extracellular matrix, mitochondrial dysfunction, chondrocyte apoptosis, and OA progression. With the continuous development of nanomaterial technology, the ROS-scavenging ability and antioxidant effects of nanomaterials are being explored, with promising results already achieved in OA treatment. However, current research on nanomaterials as ROS scavengers for OA is relatively non-uniform and includes both inorganic and functionalized organic nanomaterials. Although the therapeutic efficacy of nanomaterials has been reported to be conclusive, there is still no uniformity in the timing and potential of their use in clinical practice. This paper reviews the nanomaterials currently used as ROS scavengers for OA treatment, along with their mechanisms of action, with the aim of providing a reference and direction for similar studies, and ultimately promoting the early clinical use of nanomaterials for OA treatment. STATEMENT OF SIGNIFICANCE: Reactive oxygen species (ROS) play an important role in the pathogenesis of osteoarthritis (OA). Nanomaterials serving as promising ROS scavengers have gained increasing attention in recent years. This review provides a comprehensive overview of ROS production and regulation, as well as their role in OA pathogenesis. Furthermore, this review highlights the applications of various types of nanomaterials as ROS scavengers in OA treatment and their mechanisms of action. Finally, the challenges and future prospects of nanomaterial-based ROS scavengers in OA therapy are discussed.
Collapse
|
41
|
Rickard BP, Overchuk M, Obaid G, Ruhi MK, Demirci U, Fenton SE, Santos JH, Kessel D, Rizvi I. Photochemical Targeting of Mitochondria to Overcome Chemoresistance in Ovarian Cancer †. Photochem Photobiol 2023; 99:448-468. [PMID: 36117466 PMCID: PMC10043796 DOI: 10.1111/php.13723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022]
Abstract
Ovarian cancer is the most lethal gynecologic malignancy with a stubborn mortality rate of ~65%. The persistent failure of multiline chemotherapy, and significant tumor heterogeneity, has made it challenging to improve outcomes. A target of increasing interest is the mitochondrion because of its essential role in critical cellular functions, and the significance of metabolic adaptation in chemoresistance. This review describes mitochondrial processes, including metabolic reprogramming, mitochondrial transfer and mitochondrial dynamics in ovarian cancer progression and chemoresistance. The effect of malignant ascites, or excess peritoneal fluid, on mitochondrial function is discussed. The role of photodynamic therapy (PDT) in overcoming mitochondria-mediated resistance is presented. PDT, a photochemistry-based modality, involves the light-based activation of a photosensitizer leading to the production of short-lived reactive molecular species and spatiotemporally confined photodamage to nearby organelles and biological targets. The consequential effects range from subcytotoxic priming of target cells for increased sensitivity to subsequent treatments, such as chemotherapy, to direct cell killing. This review discusses how PDT-based approaches can address key limitations of current treatments. Specifically, an overview of the mechanisms by which PDT alters mitochondrial function, and a summary of preclinical advancements and clinical PDT experience in ovarian cancer are provided.
Collapse
Affiliation(s)
- Brittany P. Rickard
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marta Overchuk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
| | - Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson TX 95080, USA
| | - Mustafa Kemal Ruhi
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Suzanne E. Fenton
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Janine H. Santos
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - David Kessel
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Imran Rizvi
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
42
|
Wang H, Ma JX, Wu DM, Gao N, Si J, Cui BK. Identifying Bioactive Ingredients and Antioxidant Activities of Wild Sanghuangporus Species of Medicinal Fungi. J Fungi (Basel) 2023; 9:jof9020242. [PMID: 36836356 PMCID: PMC9959451 DOI: 10.3390/jof9020242] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Sanghuangporus refers to a group of rare medicinal fungi with remarkable therapeutic properties. However, current knowledge on the bioactive ingredients and antioxidant activities of different species of this genus is limited. In this study, a total of 15 wild strains from 8 species of Sanghuangporus were selected as the experimental materials for identification of the bioactive components (polysaccharide, polyphenol, flavonoid, triterpenoid, and ascorbic acid) and antioxidant activities (scavenging activities against hydroxyl, superoxide, DPPH, and ABTS radicals; superoxide dismutase activity; and ferric reducing ability of plasma). Notably, individual strains contained different levels of various indicators, among which Sanghuangporus baumii Cui 3573, S. sanghuang Cui 14419 and Cui 14441, S. vaninii Dai 9061, and S. zonatus Dai 10841 displayed the strongest activities. The correlation analysis of bioactive ingredients and antioxidant activities revealed that the antioxidant capacity of Sanghuangporus is mainly associated with the contents of flavonoid and ascorbic acid, followed by polyphenol and triterpenoid, and finally, polysaccharide. Together, the results obtained from the comprehensive and systematic comparative analyses contribute further potential resources and critical guidance for the separation, purification, and further development and utilization of bioactive agents from wild Sanghuangporus species, as well as the optimization of their artificial cultivation conditions.
Collapse
Affiliation(s)
- Hao Wang
- Institute of Microbiology, School of Ecology and Nature Conservation, Beijing Forestry University, Beijing 100083, China
| | - Jin-Xin Ma
- Institute of Microbiology, School of Ecology and Nature Conservation, Beijing Forestry University, Beijing 100083, China
| | - Dong-Mei Wu
- Xinjiang Academy of Agricultural and Reclamation Sciences/Xinjiang Production and Construction Group Key Laboratory of Crop Germplasm Enhancement and Gene Resources Utilization, Biotechnology Research Institute, Shihezi 832000, China
| | - Neng Gao
- Xinjiang Academy of Agricultural and Reclamation Sciences/Xinjiang Production and Construction Group Key Laboratory of Crop Germplasm Enhancement and Gene Resources Utilization, Biotechnology Research Institute, Shihezi 832000, China
| | - Jing Si
- Institute of Microbiology, School of Ecology and Nature Conservation, Beijing Forestry University, Beijing 100083, China
- Correspondence: (J.S.); (B.-K.C.)
| | - Bao-Kai Cui
- Institute of Microbiology, School of Ecology and Nature Conservation, Beijing Forestry University, Beijing 100083, China
- Correspondence: (J.S.); (B.-K.C.)
| |
Collapse
|
43
|
Dialynaki D, Stavropoulou A, Laskou M, Alexandraki D. The essential liaison of two copper proteins: the Cu-sensing transcription factor Mac1 and the Cu/Zn superoxide dismutase Sod1 in Saccharomyces cerevisiae. Curr Genet 2023; 69:41-53. [PMID: 36456733 DOI: 10.1007/s00294-022-01258-8] [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: 09/24/2022] [Revised: 10/30/2022] [Accepted: 11/07/2022] [Indexed: 12/04/2022]
Abstract
Although copper is an essential trace element for cell function and viability, its excess can lead to protein oxidation, DNA cleavage, and ultimate cell damage. Cells have established a variety of regulatory mechanisms to ensure copper ion homeostasis. In Saccharomyces cerevisiae, copper sensing and response to copper deficiency are regulated by the transcription factor Mac1. Our group has previously reported that in addition to copper, several chromatin proteins modulate Mac1 functionality. In this study, based on a synthetic growth deficiency phenotype, we showed that the Cu/Zn superoxide dismutase Sod1 plays an important role in Mac1 transcriptional activity, in unchallenged nutrient-rich growth conditions. Sod1 is a multipotent cytoplasmic and mitochondrial enzyme, whose main known function is to detoxify the cell from superoxide ions. It has been previously reported that Sod1 also enters the nucleus and affects the transcription of several genes, some of which are involved in copper homeostasis under Cu-depleted (Wood and Thiele in J Biol Chem 284:404-413, 2009) or only under specific oxidative stress conditions (Dong et al. Mol Cell Biol 33:4041-4050, 2013; Tsang et al. Nar Commun 8:3446, 2014). We have shown that Sod1 physically interacts with Mac1 transcription factor and is important for the transactivation as well as its DNA-binding activities. On the other hand, a constitutively active mutant of Mac1 is not affected functionally by the Sod1 ablation, pointing out that Sod1 contributes to the maintenance of the copper-unchelated state of Mac1. In conclusion, we showed that Sod1-Mac1 interaction is vital for Mac1 functionality, regardless of copper medium deficiency, in unchallenged growth conditions, and we suggest that Sod1 enzymatic activity may modify the redox state of the cysteine-rich motifs in the Mac1 DNA-binding and transactivation domains.
Collapse
Affiliation(s)
- Dimitra Dialynaki
- Department of Biology, University of Crete, Vasilika Vouton, P. O. Box 2208, 70013, Heraklion, Crete, Greece
| | - Athanasia Stavropoulou
- Department of Biology, University of Crete, Vasilika Vouton, P. O. Box 2208, 70013, Heraklion, Crete, Greece.,Computational Genomics Group, Biomedical Sciences Research Center "Alexander Fleming", 16672, Athens, Greece
| | - Maria Laskou
- Department of Biology, University of Crete, Vasilika Vouton, P. O. Box 2208, 70013, Heraklion, Crete, Greece.,NYU Langone Health, NYU Grossman School of Medicine, 435 E 30th Street, Science Building 1305, New York, NY, 10016, USA
| | - Despina Alexandraki
- Department of Biology, University of Crete, Vasilika Vouton, P. O. Box 2208, 70013, Heraklion, Crete, Greece. .,Institute of Molecular Biology & Biotechnology, Foundation for Research and Technology-Hellas, N. Plastira 100, 70013, Heraklion, Crete, Greece.
| |
Collapse
|
44
|
Cao Z, Wang H, Chen J, Zhang Y, Mo Q, Zhang P, Wang M, Liu H, Bao X, Sun Y, Zhang W, Yao Q. Silk-based hydrogel incorporated with metal-organic framework nanozymes for enhanced osteochondral regeneration. Bioact Mater 2023; 20:221-242. [PMID: 35702612 PMCID: PMC9163388 DOI: 10.1016/j.bioactmat.2022.05.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
Osteochondral defects (OCD) cannot be efficiently repaired due to the unique physical architecture and the pathological microenvironment including enhanced oxidative stress and inflammation. Conventional strategies, such as the control of implant microstructure or the introduction of growth factors, have limited functions failing to manage these complex environments. Here we developed a multifunctional silk-based hydrogel incorporated with metal-organic framework nanozymes (CuTA@SF) to provide a suitable microenvironment for enhanced OCD regeneration. The incorporation of CuTA nanozymes endowed the SF hydrogel with a uniform microstructure and elevated hydrophilicity. In vitro cultivation of mesenchymal stem cells (MSCs) and chondrocytes showed that CuTA@SF hydrogel accelerated cell proliferation and enhanced cell viability, as well as had antioxidant and antibacterial properties. Under the inflammatory environment with the stimulation of IL-1β, CuTA@SF hydrogel still possessed the potential to promote MSC osteogenesis and deposition of cartilage-specific extracellular matrix (ECM). The proteomics analysis further confirmed that CuTA@SF hydrogel promoted cell proliferation and ECM synthesis. In the full-thickness OCD model of rabbit, CuTA@SF hydrogel displayed successfully in situ OCD regeneration, as evidenced by micro-CT, histology (HE, S/O, and toluidine blue staining) and immunohistochemistry (Col I and aggrecan immunostaining). Therefore, CuTA@SF hydrogel is a promising biomaterial targeted at the regeneration of OCD. A multifunctional silk-based hydrogel incorporated with metal-organic framework nanozymes (CuTA@SF) was fabricated. CuTA@SF hydrogel has antioxidant, anti-inflammation and antibacterial capacities. Proteomics analysis confirmed that CuTA@SF hydrogel promoted cell proliferation and ECM synthesis. CuTA@SF hydrogel displayed successful osteochondral regeneration in vivo.
Collapse
Affiliation(s)
- Zhicheng Cao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Hongmei Wang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Pharmaceutical Sciences, Binzhou Medical University, 264003, Yantai, Shandong, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Yanan Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Qingyun Mo
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Po Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Mingyue Wang
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Xueyang Bao
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
- Corresponding author. School of Medicine, Southeast University, 210009, Nanjing, China.
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
- Corresponding author. Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| |
Collapse
|
45
|
Yang S, Zeng H, Jiang L, Fu C, Gao L, Zhang L, Zhang Y, Zhang X, Zhu L, Zhang F, Chen J, Huang J, Zeng Q. Melatonin reduces melanogenesis by inhibiting the paracrine effects of keratinocytes. Exp Dermatol 2023; 32:511-520. [PMID: 36620869 DOI: 10.1111/exd.14743] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/28/2022] [Accepted: 12/30/2022] [Indexed: 01/10/2023]
Abstract
Keratinocytes regulate melanogenesis in a paracrine manner. Previous studies have shown that melatonin can directly inhibit melanin production in the melanocytes. However, it is unclear whether melatonin can also indirectly regulate melanogenesis through the keratinocytes. In this study, we explored the role of melatonin in regulating keratinocyte-mediated melanogenesis using reconstructed human epidermis (RHE). Melatonin showed an inhibitory effect on melanin synthesis in this model. Furthermore, the conditioned media from melatonin-treated HaCaT cells downregulated melanogenesis-related genes, including MITF, TYR, TYRP1, DCT and RAB27A in the pigment MNT1 cells, and decreased levels of phosphorylated ERK, JNK and p38. RNA sequencing further showed that mitochondrial functions and oxidative stress pathway in the MNT1 cells were inhibited by the conditioned medium from melatonin-treated HaCaT cells. Furthermore, melatonin reduced the secretion of ET-1 and PTGS2 from HaCaT cells by inhibiting the JAK2/STAT3 signalling pathway. In conclusion, melatonin downregulates the paracrine factors ET-1 and PTGS2 in the keratinocytes by inhibiting the JAK2/STAT3 pathway, which reduces melanin production in pigment cells. Thus, melatonin has a potential therapeutic effect on skin pigmentation disorders.
Collapse
Affiliation(s)
- Siyu Yang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Hongliang Zeng
- Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, Changsha, China
| | - Ling Jiang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Chuhan Fu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lijuan Gao
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lan Zhang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yushan Zhang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaolin Zhang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lu Zhu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Fan Zhang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Jing Chen
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Jinhua Huang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Qinghai Zeng
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
46
|
Giri S, Park GH, Choi JS, Ma E, Chun KS, Joo SH. MS-5, a Naphthalene Derivative, Induces Apoptosis in Human Pancreatic Cancer BxPC-3 Cells by Modulating Reactive Oxygen Species. Biomol Ther (Seoul) 2023; 31:68-72. [PMID: 36380602 PMCID: PMC9810442 DOI: 10.4062/biomolther.2022.127] [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: 09/30/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic cancer is one of the most fatal cancers with a poor prognosis. Standard chemotherapies have proven largely ineffective because of their toxicity and the development of resistance. Therefore, there is an urgent need to develop novel therapies. In this study, we investigated the antitumor activity of MS-5, a naphthalene derivative, on BxPC-3, a human pancreatic cancer cell line. We observed that MS-5 was cytotoxic to BxPC-3 cells, as well as inhibited the growth of cells in a concentration- and time- dependent manner. Flow cytometry analysis revealed that the percentage of annexin V-positive cells increased after MS-5 treatment. We also observed cleavage of caspases and poly (ADP-ribose) polymerase, and downregulation of Bcl-xL protein. Flow cytometry analysis of intracellular levels of reactive oxygen species (ROS) and mitochondrial superoxide suggested that MS-5 induced the generation of mitochondrial superoxide while lowering the overall intracellular ROS levels. Thus, MS-5 may be potential candidate for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Suman Giri
- Department of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Gyu Hwan Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Joon-Seok Choi
- Department of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Eunsook Ma
- Department of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea,Corresponding Authors E-mail: (Joo SH), (Chun KS), Tel: +82-53-850-3614 (Joo SH), +82-53-580-6647 (Chun KS), Fax: +82-53-359-6729 (Joo SH), +82-53-580-6645 (Chun KS)
| | - Sang Hoon Joo
- Department of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea,Corresponding Authors E-mail: (Joo SH), (Chun KS), Tel: +82-53-850-3614 (Joo SH), +82-53-580-6647 (Chun KS), Fax: +82-53-359-6729 (Joo SH), +82-53-580-6645 (Chun KS)
| |
Collapse
|
47
|
Oxidative Stress and Intracranial Hypertension after Aneurysmal Subarachnoid Hemorrhage. Antioxidants (Basel) 2022; 11:antiox11122423. [PMID: 36552631 PMCID: PMC9774559 DOI: 10.3390/antiox11122423] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/25/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Intracranial hypertension is a common phenomenon in patients with aneurysmal subarachnoid hemorrhage (aSAH). Elevated intracranial pressure (ICP) plays an important role in early brain injuries and is associated with unfavorable outcomes. Despite advances in the management of aSAH, there is no consensus about the mechanisms involved in ICP increases after aSAH. Recently, a growing body of evidence suggests that oxidative stress (OS) may play a crucial role in physio-pathological changes following aSAH, which may also contribute to increased ICP. Herein, we discuss a potential relation between increased ICP and OS, and resultantly propose antioxidant mechanisms as a potential therapeutic strategy for the treatment of ICP elevation following aSAH.
Collapse
|
48
|
Damuka N, Bashetti N, Mintz A, Bansode AH, Miller M, Krizan I, Furdui C, Bhoopal B, Gollapelli KK, Shanmukha Kumar JV, Deep G, Dugan G, Cline M, Solingapuram Sai KK. [ 18F]KS1, a novel ascorbate-based ligand images ROS in tumor models of rodents and nonhuman primates. Biomed Pharmacother 2022; 156:113937. [PMID: 36411624 PMCID: PMC11017304 DOI: 10.1016/j.biopha.2022.113937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/20/2022] Open
Abstract
Over production of reactive oxygen species (ROS) caused by altered redox regulation of signaling pathways is common in many types of cancers. While PET imaging is recognized as the standard tool for cancer imaging, there are no clinically-approved PET radiotracers for ROS-imaging in cancer diagnosis and treatment. An ascorbate-based radio ligand promises to meet this urgent need. Our laboratory recently synthesized [18F] KS1, a fluoroethoxy furanose ring-containing ascorbate derivative, to track ROS in prostate tumor-bearing mice. Here we report cell uptake assays of [18F]KS1 with different ROS-regulating agents, PET imaging in head and neck squamous cell carcinoma (HNSCC) mice, and doxorubicin-induced rats; PET imaging in healthy and irradiated hepatic tumor-bearing rhesus to demonstrate its translational potential. Our preliminary evaluations demonstrated that KS1 do not generate ROS in tumor cells at tracer-level concentrations and tumor-killing properties at pharmacologic doses. [18F]KS1 uptake was low in HNSCC pretreated with ROS blockers, and high with ROS inducers. Tumors in high ROS-expressing SCC-61 took up significantly more [18F]KS1 than rSCC-61 (low-ROS expressing HNSCC); high uptake in doxorubicin-treated rats compared to saline-treated controls. Rodent biodistribution and PET imaging of [18F]KS1 in healthy rhesus monkeys demonstrated its favorable safety, pharmacokinetic properties with excellent washout profile, within 3.0 h of radiotracer administration. High uptake of [18F]KS1 in liver tumor tissues of the irradiated hepatic tumor-bearing monkey showed target selectivity. Our strong data in vitro, in vivo, and ex vivo here supports the high translational utility of [18F]KS1 to image ROS.
Collapse
Affiliation(s)
- Naresh Damuka
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Nagaraju Bashetti
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Andhra Pradesh, India
| | - Akiva Mintz
- Department of Radiology, Columbia University, New York, NY, United States
| | - Avinash H Bansode
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Mack Miller
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Ivan Krizan
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Cristina Furdui
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Bhuvanachandra Bhoopal
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | | | - J V Shanmukha Kumar
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Andhra Pradesh, India
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Greg Dugan
- Department of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Mark Cline
- Department of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | | |
Collapse
|
49
|
Khan SU, Fatima K, Aisha S, Hamza B, Malik F. Redox balance and autophagy regulation in cancer progression and their therapeutic perspective. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:12. [PMID: 36352310 DOI: 10.1007/s12032-022-01871-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/30/2022] [Indexed: 11/10/2022]
Abstract
Cellular ROS production participates in various cellular functions but its accumulation decides the cell fate. Malignant cells have higher levels of ROS and active antioxidant machinery, a characteristic hallmark of cancer with an outcome of activation of stress-induced pathways like autophagy. Autophagy is an intracellular catabolic process that produces alternative raw materials to meet the energy demand of cells and is influenced by the cellular redox state thus playing a definite role in cancer cell fate. Since damaged mitochondria are the main source of ROS in the cell, however, cancer cells remove them by upregulating the process of mitophagy which is known to play a decisive role in tumorigenesis and tumor progression. Chemotherapy exploits cell machinery which results in the accumulation of toxic levels of ROS in cells resulting in cell death by activating either of the pathways like apoptosis, necrosis, ferroptosis or autophagy in them. So understanding these redox and autophagy regulations offers a promising method to design and develop new cancer therapies that can be very effective and durable for years. This review will give a summary of the current therapeutic molecules targeting redox regulation and autophagy for the treatment of cancer. Further, it will highlight various challenges in developing anticancer agents due to autophagy and ROS regulation in the cell and insights into the development of future therapies.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Sanat Nagar, Ghaziabad, 201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Sanat Nagar, Ghaziabad, 201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, Jammu and Kashmir, India
| | - Baseerat Hamza
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, Jammu and Kashmir, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, 190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Sanat Nagar, Ghaziabad, 201002, India.
| |
Collapse
|
50
|
Hu H, Xu Q, Mo Z, Hu X, He Q, Zhang Z, Xu Z. New anti-cancer explorations based on metal ions. J Nanobiotechnology 2022; 20:457. [PMID: 36274142 PMCID: PMC9590139 DOI: 10.1186/s12951-022-01661-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/03/2022] [Indexed: 12/07/2022] Open
Abstract
Due to the urgent demand for more anti-cancer methods, the new applications of metal ions in cancer have attracted increasing attention. Especially the three kinds of the new mode of cell death, including ferroptosis, calcicoptosis, and cuproptosis, are of great concern. Meanwhile, many metal ions have been found to induce cell death through different approaches, such as interfering with osmotic pressure, triggering biocatalysis, activating immune pathways, and generating the prooxidant effect. Therefore, varieties of new strategies based on the above approaches have been studied and applied for anti-cancer applications. Moreover, many contrast agents based on metal ions have gradually become the core components of the bioimaging technologies, such as MRI, CT, and fluorescence imaging, which exhibit guiding significance for cancer diagnosis. Besides, the new nano-theranostic platforms based on metal ions have experimentally shown efficient response to endogenous and exogenous stimuli, which realizes simultaneous cancer therapy and diagnosis through a more controlled nano-system. However, most metal-based agents have still been in the early stages, and controlled clinical trials are necessary to confirm or not the current expectations. This article will focus on these new explorations based on metal ions, hoping to provide some theoretical support for more anti-cancer ideas.
Collapse
Affiliation(s)
- Han Hu
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Qi Xu
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Zhimin Mo
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Xiaoxi Hu
- College of Petroleum and Chemical Engineering, Beibu Gulf University, Qinzhou, 535011, China
| | - Qianyuan He
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan, 430062, Hubei, China.
| | - Zhanjie Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zushun Xu
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science and Engineering, Hubei University, Wuhan, 430062, Hubei, China.
| |
Collapse
|