1
|
Wang T, Cao J, Hou Z, Zhang Q. Serum inflammation-related proteins in a acute compartment syndrome rat model. Sci Rep 2025; 15:91. [PMID: 39747444 PMCID: PMC11696460 DOI: 10.1038/s41598-024-83796-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/05/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
We aim to explore variations of serum inflammation-related proteins in an acute compartment syndrome (ACS) rat model. We collected serum from 25 healthy Sprague-Dawley rats (control group, CG) and 50 rats with tibial fractures, including 25 rats with ACS (ACS group, AG), and 25 rats without ACS (fracture group, FG). Ten samples per group were randomly chosen for proximity extension assay analysis of 92 inflammation-related proteins, and all samples were verified by enzyme-linked immunosorbent assays. Receiver-operating characteristic curve analysis was used to identify the diagnostic ability and cut-off values. Our findings showed that the levels of Il6 and Prdx5 in the FG and Il6, Prdx5, Dctn2, and Plin1 in the AG, were significantly higher than those in the CG. Notably, compared with the FG, high expression of Prdx5, Dctn2, and Plin1 was observed in the AG. Additionally, we identified 58.8764, 14.023, and 31.8730 pg/ml as the cut-off values of Prdx5, Dctn2, and Plin1 to predict ACS in rats. Similarly, the cut-off values of Il6, Prdx5, Dctn2, and Plin1 to predict ACS in healthy rats were 10.6783, 766.5879, 12.5627, and 14.3623 pg/ml, respectively. Furthermore, the combination of these proteins had the highest diagnostic accuracy. We identified Prdx5, Dctn2, and Plin1 as potential biomarkers of ACS compared with fracture in rats and revealed that combination of Il6, Prdx5, Dctn2, and Plin1 had the highest diagnostic accuracy to predict ACS compared with the healthy condition. Furthermore, the cut-off values for these biomarkers were determined, providing a new method to rapidly assess the risk of ACS and manage early targeted interventions.
Collapse
Affiliation(s)
- Tao Wang
- Department of Lower Limb Trauma, Beijing Jishuitan Hospital, Guizhou Hospital, Baiyun District, Guiyang, Guizhou, China
| | - Jingcheng Cao
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
- Orthopaedic Research Institute of Hebei Province, Shijiazhuang, Hebei, P.R. China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China.
- Orthopaedic Research Institute of Hebei Province, Shijiazhuang, Hebei, P.R. China.
- Department of Anesthesiology, Hebei Children's Hospital, No133 Jianhua South Street, Shijiazhuang, Hebei province, China.
| | - Qi Zhang
- Department of Anesthesiology, Children's hospital of Hebei Province, Shijiazhuang, Hebei province, China.
- Department of Anesthesiology, Hebei Children's Hospital, No133 Jianhua South Street, Shijiazhuang, Hebei province, China.
| |
Collapse
|
2
|
Wang R, Pan Y, Zhang L, Wang J, Ni J, Ding Y, Wang S, Yin J, Ding L, Ran X, Fan S, Sun Q, Tan SY, Koeffler HP, Li J, Mi Y, Chen YQ. Prebiotic stachyose inhibits PRDX5 activity and castration-resistant prostate cancer development. Int J Biol Macromol 2024; 278:134844. [PMID: 39168191 DOI: 10.1016/j.ijbiomac.2024.134844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/22/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Stachyose (STA) is a prebiotic with poor oral bioavailability. In this study, we developed stachyose caproate (C6-STA), as a novel STA derivative, to demonstrate its high adsorption rate via oral administration. Pharmacokinetic analysis reveals that after absorption, the STA derived from C6-STA reaches its highest peak in the blood, liver, and kidney at 20 min, 30 min, and 12-24 h, with approximate levels of 1200 μg/mL, 0.14 μg/mL, and 0.2-0.3 μg/mL, respectively. In addition, the accumulation of STA in prostate tissues of mice with castration-resistant prostate cancer (CRPC) (1.75 μg/mg) is 10-fold higher than that in normal prostate tissues (0.14 μg/mg). The analysis also reveals that C6-STA has t1/2 of 12.8 h and Tmax of 0.25 h, indicating that it has the potential to be used as a promising drug in clinical practice. The toxicological evaluation shows no obvious side effects of C6-STA in mice administered with a 0.2 g/kg intragastric dose. Pharmacodynamic analysis and mechanism investigation of C6-STA show its ability to inhibit peroxiredoxin 5 (PRDX5) enzyme activity, disrupt PRDX5-nuclear factor erythroid 2-related factor 2 (NRF2) interaction, and decrease NAD(P)H quinone dehydrogenase 1 (NQO1) levels. NQO1 decrease further causes the accumulation of quinone radicals, which ultimately leads to the apoptosis of LNCaP cell-derived drug-tolerant persister (DTP) cells and slows CRPC progression. Our study discovered the anti-tumor activity of stachyose and shows that prebiotics have biological functions in vivo besides in the gut. Further investigation of C6-STA, especially in CRPC patients, is warranted.
Collapse
Affiliation(s)
- Rong Wang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yu Pan
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lan Zhang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jun Wang
- First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China; Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Jiang Ni
- Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Yang Ding
- College of Pharmacy, Pharmaceutical Series, China Pharmaceutical University, Nanjing, China
| | - Shaopeng Wang
- Jiangnan University Medical Center, Jiangnan University, Wuxi, China
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology & School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Lingwen Ding
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xuebin Ran
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Shuangyi Fan
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Qiaoyang Sun
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
| | - Soo Yong Tan
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; Division of Hematology/Oncology, Cedars-Sinai Medical Center, UCLA School of Medicine, CA, Los Angeles, USA
| | - Jie Li
- First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.
| | - Yuanyuan Mi
- Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, China.
| | - Yong Q Chen
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China.
| |
Collapse
|
3
|
Sun J, Zhang Z, Xia B, Yao T, Ge F, Yan F. circ_0114866 promotes the progression and EMT of non-small cell lung cancer via miR-653-5p/MYL6B axis. Heliyon 2024; 10:e37062. [PMID: 39281569 PMCID: PMC11402247 DOI: 10.1016/j.heliyon.2024.e37062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/20/2023] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/18/2024] Open
Abstract
Background Non-small-cell lung cancer (NSCLC) is the most prevalent form of lung cancer. Circular RNA (circRNA) has emerged as a key player in the development of NSCLC by acting as miRNA sponges. However, the precise role of circ_0114866 in regulating NSCLC process is yet to be elucidated. Methods The expression of circ_0114866, miR-653-5p, and MYL6B were assessed by qPCR. Cell viability, proliferation, invasion, and migration were investigated using CCK-8, colony formation, Transwell, and wound healing assays. The protein levels of MYL6B, MMP-2, N-cadherin, E-cadherin, and vimentin were evaluated through Western blot analysis. Xenograft tumor model were selected to analyze the impact of circ_0114866 on NSCLC tumor growth. Through circBank or Starbase databases, the binding interactions between miR-653-5p and circ_0114866 or MYL6B were predicted. Subsequently, these interactions were verified by dual-luciferase reporter assay. Results The expression of circ_0114866 and MYL6B were clearly elevated, while miR-653-5p expression was notably reduced in NSCLC tissues and cells. Notably, circ_0114866 knockdown obviously suppressed the proliferation, metastasis, and EMT process in NSCLC cells. Additionally, circ_0114866 functioned as a sponge for miR-653-5p, leading to an increase in MYL6B expression by absorbing miR-653-5p. Furthermore, the inhibitory effects on biological behaviors and EMT process of NSCLC cells induced by circ_0114866 knockdown were reversed by miR-653-5p inhibitor. Moreover, in vivo experiments demonstrated that silencing circ_0114866 resulted in a repression of tumor growth. Conclusion Our findings indicate that circ_0114866 knockdown upregulated MYL6B transcription by sponging miR-653-5p, leading to hinder the progression and EMT process of NSCLC.
Collapse
Affiliation(s)
- Jinpeng Sun
- Department of General Surgery Ward,Cangzhou Hospital of Integrated TCM-WM·Hebei China
| | - Zhenshan Zhang
- Department of Medical Oncology, Cangzhou Hospital of Integrated TCM-WM·Hebei, China
| | - Binghui Xia
- Department of General Surgery Ward,Cangzhou Hospital of Integrated TCM-WM·Hebei China
| | - Tianyu Yao
- Department of Cardiology, Cangzhou Hospital of Integrated TCM-WM·Hebei, China
| | - Fengyue Ge
- Department of Functional Examination, Cangzhou Hospital of Integrated TCM-WM·Hebei, China
| | - Fengmei Yan
- Department of Endoscopic Diagnosis and Treatment Center, Cangzhou Hospital of Integrated TCM-WM·Hebei, China
| |
Collapse
|
4
|
Abad C, Pinal-Fernandez I, Guillou C, Bourdenet G, Drouot L, Cosette P, Giannini M, Debrut L, Jean L, Bernard S, Genty D, Zoubairi R, Remy-Jouet I, Geny B, Boitard C, Mammen A, Meyer A, Boyer O. IFNγ causes mitochondrial dysfunction and oxidative stress in myositis. Nat Commun 2024; 15:5403. [PMID: 38926363 PMCID: PMC11208592 DOI: 10.1038/s41467-024-49460-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/28/2023] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are severe autoimmune diseases with poorly understood pathogenesis and unmet medical needs. Here, we examine the role of interferon γ (IFNγ) using NOD female mice deficient in the inducible T cell co-stimulator (Icos), which have previously been shown to develop spontaneous IFNγ-driven myositis mimicking human disease. Using muscle proteomic and spatial transcriptomic analyses we reveal profound myofiber metabolic dysregulation in these mice. In addition, we report muscle mitochondrial abnormalities and oxidative stress in diseased mice. Supporting a pathogenic role for oxidative stress, treatment with a reactive oxygen species (ROS) buffer compound alleviated myositis, preserved muscle mitochondrial ultrastructure and respiration, and reduced inflammation. Mitochondrial anomalies and oxidative stress were diminished following anti-IFNγ treatment. Further transcriptomic analysis in IIMs patients and human myoblast in vitro studies supported the link between IFNγ and mitochondrial dysfunction observed in mice. These results suggest that mitochondrial dysfunction, ROS and inflammation are interconnected in a self-maintenance loop, opening perspectives for mitochondria therapy and/or ROS targeting drugs in myositis.
Collapse
Affiliation(s)
- Catalina Abad
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - Iago Pinal-Fernandez
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clement Guillou
- Univ Rouen Normandie, Inserm US 51, CNRS UAR 2026, HeRacLeS PISSARO, F-76000, Rouen, France
| | - Gwladys Bourdenet
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - Laurent Drouot
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - Pascal Cosette
- Univ Rouen Normandie, Inserm US 51, CNRS UAR 2026, HeRacLeS PISSARO, F-76000, Rouen, France
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000, Rouen, France
| | - Margherita Giannini
- Translational Medicine Federation of Strasbourg, Team 3072, Faculty of Medicine, University of Strasbourg, Strasbourg, France
- Unité exploration fonctionnelle musculaire-service de physiologie, Centre National de Référence des Maladies Auto-Immunes Systémiques Rares de l'Est et du Sud-Ouest -Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Lea Debrut
- Translational Medicine Federation of Strasbourg, Team 3072, Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Laetitia Jean
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - Sophie Bernard
- Univ Rouen Normandie, Inserm US51, CNRS UAR2026, HeRacLeS PRIMACEN, F-76000, Rouen, France
| | - Damien Genty
- CHU Rouen, Department of Pathology, F-76000, Rouen, France
| | - Rachid Zoubairi
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - Isabelle Remy-Jouet
- Univ Rouen Normandie, Inserm, UMR1096, BOSS facility, F-76000, Rouen, France
| | - Bernard Geny
- Translational Medicine Federation of Strasbourg, Team 3072, Faculty of Medicine, University of Strasbourg, Strasbourg, France
- Unité exploration fonctionnelle musculaire-service de physiologie, Centre National de Référence des Maladies Auto-Immunes Systémiques Rares de l'Est et du Sud-Ouest -Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Christian Boitard
- Cochin Institute, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Andrew Mammen
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alain Meyer
- Translational Medicine Federation of Strasbourg, Team 3072, Faculty of Medicine, University of Strasbourg, Strasbourg, France
- Unité exploration fonctionnelle musculaire-service de physiologie, Centre National de Référence des Maladies Auto-Immunes Systémiques Rares de l'Est et du Sud-Ouest -Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Olivier Boyer
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France.
- CHU Rouen, Department of Immunology and Biotherapy, F-76000, Rouen, France.
| |
Collapse
|
5
|
Chen H, Han Z, Su J, Song X, Ma Q, Lin Y, Ran Z, Li X, Mou R, Wang Y, Li D. Ferroptosis and hepatocellular carcinoma: the emerging role of lncRNAs. Front Immunol 2024; 15:1424954. [PMID: 38846953 PMCID: PMC11153672 DOI: 10.3389/fimmu.2024.1424954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 06/09/2024] Open
Abstract
Hepatocellular carcinoma is the most common form of primary liver cancer and poses a significant challenge to the medical community because of its high mortality rate. In recent years, ferroptosis, a unique form of cell death, has garnered widespread attention. Ferroptosis, which is characterized by iron-dependent lipid peroxidation and mitochondrial alterations, is closely associated with the pathological processes of various diseases, including hepatocellular carcinoma. Long non-coding RNAs (lncRNAs), are a type of functional RNA, and play crucial regulatory roles in a variety of biological processes. In this manuscript, we review the regulatory roles of lncRNAs in the key aspects of ferroptosis, and summarize the research progress on ferroptosis-related lncRNAs in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Haoran Chen
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zhongyu Han
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Junyan Su
- The First People’s Hospital of Longquanyi District, Chengdu, China
| | - Xuanliang Song
- The First People’s Hospital of Longquanyi District, Chengdu, China
| | - Qingquan Ma
- The First People’s Hospital of Longquanyi District, Chengdu, China
| | - Yumeng Lin
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zijin Ran
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Xueping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rongkun Mou
- Department of General Surgery, The Third Hospital of Mianyang, Mianyang, China
| | - Yi Wang
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Dongxuan Li
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| |
Collapse
|
6
|
Jiang H, Song D, Zhou X, Chen F, Yu Q, Ren L, Dai Q, Zeng M. Maresin1 ameliorates MSU crystal-induced inflammation by upregulating Prdx5 expression. Mol Med 2023; 29:158. [PMID: 37996809 PMCID: PMC10668345 DOI: 10.1186/s10020-023-00756-w] [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] [Academic Contribution Register] [Received: 06/20/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Maresin1 (MaR1) is a potent lipid mediator that exhibits significant anti-inflammatory activity in the context of several inflammatory diseases. A previous study reported that MaR1 could suppress MSU crystal-induced peritonitis in mice. To date, the molecular mechanism by which MaR1 inhibits MSU crystal-induced inflammation remains poorly understood. METHODS Mousebone marrow-derived macrophages (BMDMs) were pretreated with MaR1 and then stimulated with FAs (palmitic, C16:0 and stearic, C18:0) plus MSU crystals (FAs + MSUc). In vivo, the effects of MaR1 treatment or Prdx5 deficiency on MSUc induced peritonitis and arthritis mouse models were evaluated. RESULTS The current study indicated that MaR1 effectively suppressed MSUc induced inflammation in vitro and in vivo. MaR1 reversed the decrease in Prdx5 mRNA and protein levels induced by FAs + MSUc. Further assays demonstrated that MaR1 acceleratedPrdx5 expression by regulating the Keap1-Nrf2 signaling axis. Activation of AMPK by Prdx5 improved homeostasis of the TXNIP and TRX proteins and alleviated mitochondrial fragmentation. In addition, Prdx5 overexpression inhibited the expression of CPT1A, a key enzyme for fatty acid oxidation (FAO). Prdx5 protected against defects in FA + MSUc induced FAO and the urea cycle. CONCLUSION MaR1 treatment effectively attenuated MSUc induced inflammation by upregulating Prdx5 expression. Our study provides a new strategy by which Prdx5 may help prevent acute gout attacks.
Collapse
Affiliation(s)
- Hui Jiang
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, 1# South Maoyuan Road, Nanchong, 637001, Sichuan Province, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 234# Fujiang Road, Nanchong, 637001, Sichuan Province, China
| | - DianZe Song
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, 1# South Maoyuan Road, Nanchong, 637001, Sichuan Province, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 234# Fujiang Road, Nanchong, 637001, Sichuan Province, China
- Medical Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of North Sichuan Medical College, 1# South Maoyuan Road, Nanchong, 637001, Sichuan, China
| | - Xiaoqin Zhou
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, 1# South Maoyuan Road, Nanchong, 637001, Sichuan Province, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 234# Fujiang Road, Nanchong, 637001, Sichuan Province, China
| | - Feng Chen
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, 1# South Maoyuan Road, Nanchong, 637001, Sichuan Province, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 234# Fujiang Road, Nanchong, 637001, Sichuan Province, China
| | - Qingqing Yu
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, 1# South Maoyuan Road, Nanchong, 637001, Sichuan Province, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 234# Fujiang Road, Nanchong, 637001, Sichuan Province, China
- Medical Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of North Sichuan Medical College, 1# South Maoyuan Road, Nanchong, 637001, Sichuan, China
| | - Long Ren
- The Fifth People's Hospital of Nanchong City, 21# Bajiao Street, Nanchong, 637100, Sichuan, China
| | - Qian Dai
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, 1# South Maoyuan Road, Nanchong, 637001, Sichuan Province, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 234# Fujiang Road, Nanchong, 637001, Sichuan Province, China
| | - Mei Zeng
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, 1# South Maoyuan Road, Nanchong, 637001, Sichuan Province, China.
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 234# Fujiang Road, Nanchong, 637001, Sichuan Province, China.
- Medical Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of North Sichuan Medical College, 1# South Maoyuan Road, Nanchong, 637001, Sichuan, China.
- The Fifth People's Hospital of Nanchong City, 21# Bajiao Street, Nanchong, 637100, Sichuan, China.
| |
Collapse
|
7
|
Bruschi M, Biancucci F, Masini S, Piacente F, Ligi D, Bartoccini F, Antonelli A, Mannello F, Bruzzone S, Menotta M, Fraternale A, Magnani M. The influence of redox modulation on hypoxic endothelial cell metabolic and proteomic profiles through a small thiol-based compound tuning glutathione and thioredoxin systems. Biofactors 2023; 49:1205-1222. [PMID: 37409789 DOI: 10.1002/biof.1988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/10/2023] [Accepted: 06/16/2023] [Indexed: 07/07/2023]
Abstract
Reduction in oxygen levels is a key feature in the physiology of the bone marrow (BM) niche where hematopoiesis occurs. The BM niche is a highly vascularized tissue and endothelial cells (ECs) support and regulate blood cell formation from hematopoietic stem cells (HSCs). While in vivo studies are limited, ECs when cultured in vitro at low O2 (<5%), fail to support functional HSC maintenance due to oxidative environment. Therefore, changes in EC redox status induced by antioxidant molecules may lead to alterations in the cellular response to hypoxia likely favoring HSC self-renewal. To evaluate the impact of redox regulation, HUVEC, exposed for 1, 6, and 24 h to 3% O2 were treated with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152). Metabolomic analyses revealed that I-152 increased glutathione levels and influenced the metabolic profiles interconnected with the glutathione system and the redox couples NAD(P)+/NAD(P)H. mRNA analysis showed a lowered gene expression of HIF-1α and VEGF following I-152 treatment whereas TRX1 and 2 were stimulated. Accordingly, the proteomic study revealed the redox-dependent upregulation of thioredoxin and peroxiredoxins that, together with the glutathione system, are the main regulators of intracellular ROS. Indeed, a time-dependent ROS production under hypoxia and a quenching effect of the molecule were evidenced. At the secretome level, the molecule downregulated IL-6, MCP-1, and PDGF-bb. These results suggest that redox modulation by I-152 reduces oxidative stress and ROS level in hypoxic ECs and may be a strategy to fine-tune the environment of an in vitro BM niche able to support functional HSC maintenance.
Collapse
Affiliation(s)
- Michela Bruschi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Federica Biancucci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Sofia Masini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Francesco Piacente
- Department of Experimental Medicine, Section of Biochemistry, and CEBR, University of Genoa, Genoa, GE, Italy
| | - Daniela Ligi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Francesca Bartoccini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Antonella Antonelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Ferdinando Mannello
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, and CEBR, University of Genoa, Genoa, GE, Italy
- IRCCS, Ospedale Policlinico San Martino, Genoa, GE, Italy
| | - Michele Menotta
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Alessandra Fraternale
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, Italy
| |
Collapse
|
8
|
Jambrovics K, Póliska S, Scholtz B, Uray IP, Balajthy Z. ATO Increases ROS Production and Apoptosis of Cells by Enhancing Calpain-Mediated Degradation of the Cancer Survival Protein TG2. Int J Mol Sci 2023; 24:10938. [PMID: 37446117 DOI: 10.3390/ijms241310938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/09/2023] [Revised: 05/14/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Transglutaminase 2 (TG2) is a critical cancer cell survival factor that activates several signalling pathways to foster drug resistance, cancer stem cell survival, metastasis, inflammation, epithelial-mesenchymal transition, and angiogenesis. All-trans retinoic acid (ATRA) and chemotherapy have been the standard treatments for acute promyelocytic leukaemia (APL), but clinical studies have shown that arsenic trioxide (ATO), alone or in combination with ATRA, can improve outcomes. ATO exerts cytotoxic effects in a variety of ways by inducing oxidative stress, genotoxicity, altered signal transduction, and/or epigenetic modification. In the present study, we showed that ATO increased ROS production and apoptosis ratios in ATRA-differentiated NB4 leukaemia cells, and that these responses were enhanced when TG2 was deleted. The combined ATRA + ATO treatment also increased the amount of nuclear factor erythroid 2-related factor 2 (NRF2) transcription factor, an adaptive regulator of the cellular oxidative stress response, and calpain proteolytic activity, resulting in TG2 degradation and the reduced survival of WT leukaemia cells. We further showed that the induced TG2 protein expression was degraded in the MCF-7 epithelial cell line and primary peripheral blood mononuclear cells upon ATO treatment, thereby sensitising these cell types to apoptotic signals.
Collapse
Affiliation(s)
- Károly Jambrovics
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Szilárd Póliska
- Genomic Medicine and Bioinformatic Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Beáta Scholtz
- Genomic Medicine and Bioinformatic Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Iván P Uray
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- Department of Clinical Oncology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Zoltán Balajthy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| |
Collapse
|
9
|
Liu Y, Wang P, Hu W, Chen D. New insights into the roles of peroxiredoxins in cancer. Biomed Pharmacother 2023; 164:114896. [PMID: 37210897 DOI: 10.1016/j.biopha.2023.114896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/23/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023] Open
Abstract
Oxidative stress is one of the hallmarks of cancer. Tumorigenesis and progression are accompanied by elevated reactive oxygen species (ROS) levels and adaptive elevation of antioxidant expression levels. Peroxiredoxins (PRDXs) are among the most important antioxidants and are widely distributed in a variety of cancers. PRDXs are involved in the regulation of a variety of tumor cell phenotypes, such as invasion, migration, epithelial-mesenchymal transition (EMT) and stemness. PRDXs are also associated with tumor cell resistance to cell death, such as apoptosis and ferroptosis. In addition, PRDXs are involved in the transduction of hypoxic signals in the TME and in the regulation of the function of other cellular components of the TME, such as cancer-associated fibroblasts (CAFs), natural killer (NK) cells and macrophages. This implies that PRDXs are promising targets for cancer treatment. Of course, further studies are needed to realize the clinical application of targeting PRDXs. In this review, we highlight the role of PRDXs in cancer, summarizing the basic features of PRDXs, their association with tumorigenesis, their expression and function in cancer, and their relationship with cancer therapeutic resistance.
Collapse
Affiliation(s)
- Yan Liu
- First Department of Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning, China
| | - Pu Wang
- Department of Emergency, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning, China
| | - Weina Hu
- Department of General Practice, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning, China.
| | - Da Chen
- Department of Emergency, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning, China.
| |
Collapse
|
10
|
Thapa P, Jiang H, Ding N, Hao Y, Alshahrani A, Wei Q. The Role of Peroxiredoxins in Cancer Development. BIOLOGY 2023; 12:666. [PMID: 37237480 PMCID: PMC10215932 DOI: 10.3390/biology12050666] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/04/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023]
Abstract
Peroxiredoxins (Prxs) are antioxidant enzymes with ubiquitous expression in human tissues. Prxs are expressed in archaea, bacteria, and eukaryota, often in multiple isoforms. Because of their abundant expression in different cellular organelles and extraordinary sensitivity to H2O2, Prxs are among the first defenses against oxidative stress. Prxs undergo reversible oxidation to disulfides, and some family members perform chaperone or phospholipase functions upon further oxidation. Prxs are upregulated in cancer cells. Research has suggested that Prxs can function as tumor promoters in various cancers. The major objective of this review is to summarize novel findings regarding the roles of Prxs in common cancer types. Prxs have been shown to influence differentiation of inflammatory cells and fibroblasts, remodeling of extracellular matrix, and regulation of stemness. Since aggressive cancer cells have higher intracellular levels of ROS that they can utilize to proliferate and metastasize compared to normal cells, it is critical that we understand the regulation and functions of primary antioxidants such as Prxs. These small but mighty proteins could prove to be key for improving cancer therapeutics and patient survival.
Collapse
Affiliation(s)
- Pratik Thapa
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
| | - Hong Jiang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
| | - Na Ding
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
| | - Yanning Hao
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
| | - Aziza Alshahrani
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
| | - Qiou Wei
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
- Markey Cancer Center, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536, USA
| |
Collapse
|
11
|
Holvoet P. Noncoding RNAs Controlling Oxidative Stress in Cancer. Cancers (Basel) 2023; 15:cancers15041155. [PMID: 36831498 PMCID: PMC9954372 DOI: 10.3390/cancers15041155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/16/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Mitochondria in cancer cells tend to overproduce reactive oxygen species (ROS), inducing a vicious cycle between mitochondria, ROS, genomic instability, and cancer development. The first part of this review deals with the role of noncoding RNAs in regulating mitochondrial ROS production and the expression of antioxidants in cancer cells, preventing the increase of ROS in the tumor microenvironment. In addition, cytotoxic T and natural killer cells release high levels of ROS, inducing cell death, while anti-immune regulatory T cells, tumor-associated M2 macrophages, and myeloid-derived suppressor cells, at least at the initial stage of tumor growth, release low levels of ROS supporting tumor growth. Therefore, this review's second part deals with noncoding RNAs' role in regulating the metabolic reprogramming of immune cells about ROS release. Furthermore, the enrichment of noncoding RNAs in microvesicles allows communication between cell types in a tumor and between a tumor and tumor-adjacent tissues. Therefore, the third part illustrates how noncoding RNA-containing microvesicles secreted by mesenchymal stem cells and primary tumor cells may primarily aid the shift of immune cells to a pro-oncogenic phenotype. Conversely, microvesicles released by tumor-adjacent tissues may have the opposite effect. Our review reveals that a specific noncoding RNA may affect oxidative stress by several mechanisms, which may have opposite effects on tumor growth. Furthermore, they may be involved in mechanisms other than regulating oxidative stress, which may level out their effects on oxidative stress and tumor growth. In addition, several noncoding RNAs might share a specific function, making it very unlikely that intervening with only one of these noncoding RNAs will block this particular mechanism. Overall, further validation of the interaction between noncoding RNAs about cancer types and stages of tumor development is warranted.
Collapse
Affiliation(s)
- Paul Holvoet
- Division of Experimental Cardiology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
12
|
QIAN SITONG, FANG YING, YAO CHENGYUN, WANG YONGSHENG, ZHANG ZHI, WANG XIAOHUA, GAO JIN, FENG YONG, SUN LEI, ZOU RUNYUE, ZHOU GUOREN, YE JINJUN, XIA RUIXUE, XIA HONGPING. The synergistic effects of PRDX5 and Nrf2 on lung cancer progression and drug resistance under oxidative stress in the zebrafish models. Oncol Res 2023; 30:53-64. [PMID: 37305326 PMCID: PMC10208055 DOI: 10.32604/or.2022.026302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/29/2022] [Accepted: 11/15/2022] [Indexed: 01/06/2023] Open
Abstract
Previous studies have shown that PRDX5 and Nrf2 are antioxidant proteins related to abnormal reactive oxidative species (ROS). PRDX5 and Nrf2 play a critical role in the progression of inflammations and tumors. The combination of PRDX5 and Nrf2 was examined by Co-immunoprecipitation, western blotting and Immunohistochemistry. H2O2 was applied to affect the production of ROS and induced multi-resistant protein 1 (MRP1) expression in NSCLC cells. The zebrafish models mainly investigated the synergistic effects of PRDX5 and Nrf2 on lung cancer drug resistance under oxidative stress. We showed that PRDX5 and Nrf2 form a complex and significantly increase the NSCLC tissues compared to adjacent tissues. The oxidative stress improved the combination of PRDX5 and Nrf2. We demonstrated that the synergy between PRDX5 and Nrf2 is positively related to the proliferation and drug resistance of NSCLC cells in the zebrafish models. In conclusion, our data indicated that PRDX5 could bind to Nrf2 and has a synergistic effect with Nrf2. Meanwhile, in the zebrafish models, PRDX5 and Nrf2 have significant regulatory impacts on lung cancer progression and drug resistance activities under oxidative stress.
Collapse
Affiliation(s)
- SITONG QIAN
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
- School of Life Sciences, Nanjing Normal University, Nanjing, 210046, China
| | - YING FANG
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - CHENGYUN YAO
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - YONGSHENG WANG
- Department of Respiratory Medicine, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, 210008, China
| | - ZHI ZHANG
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - XIAOHUA WANG
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - JIN GAO
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - YONG FENG
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - LEI SUN
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - RUNYUE ZOU
- School of Life Sciences, Nanjing Normal University, Nanjing, 210046, China
| | - GUOREN ZHOU
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - JINJUN YE
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
| | - RUIXUE XIA
- Medical College of Henan University & Henan University Huaihe Hospital, Kaifeng, 475000, China
| | - HONGPING XIA
- Zhongda Hospital, School of Medicine & Advanced Institute for Life and Health, Southeast University, Nanjing, 210009, China
- Department of Pathology, Nanjing Drum Tower Hospital & Drum Tower Clinical College & School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
13
|
Potential Cytoprotective and Regulatory Effects of Ergothioneine on Gene Expression of Proteins Involved in Erythroid Adaptation Mechanisms and Redox Pathways in K562 Cells. Genes (Basel) 2022; 13:genes13122368. [PMID: 36553634 PMCID: PMC9778224 DOI: 10.3390/genes13122368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/20/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
This study aimed to establish the importance of ergothioneine (ERT) in the erythroid adaptation mechanisms by appraising the expression levels of redox-related genes associated with the PI3K/AKT/FoxO3 and Nrf2-ARE pathways using K562 cells induced to erythroid differentiation and H2O2-oxidative stress. Cell viability and gene expression were evaluated. Two concentrations of ERT were assessed, 1 nM (C1) and 100 µM (C2), with and without stress induction (100 µM H2O2). Assessments were made in three periods of the cellular differentiation process (D0, D2, and D4). The C1 treatment promoted the induction of FOXO3 (D0 and 2), PSMB5, and 6 expressions (D4); C1 + H2O2 treatment showed the highest levels of NRF2 transcripts, KEAP1 (D0), YWHAQ (D2 and 4), PSMB5 (D2) and PSMB6 (D4); and C2 + H2O2 (D2) an increase in FOXO3 and MST1 expression, with a decrease of YWHAQ and NRF2 was observed. in C2 + H2O2 (D2) an increase in FOXO3 and MST1, with a decrease in YWHAQ and NRF2 was observed All ERT treatments increased gamma-globin expression. Statistical multivariate analyzes highlighted that the Nrf2-ARE pathway presented a greater contribution in the production of PRDX1, SOD1, CAT, and PSBM5 mRNAs, whereas the PI3K/AKT/FoxO3 pathway was associated with the PRDX2 and TRX transcripts. In conclusion, ERT presented a cytoprotective action through Nrf2 and FoxO3, with the latter seeming to contribute to erythroid proliferation/differentiation.
Collapse
|
14
|
Proteomic Analysis of Human Follicular Fluid Reveals the Pharmacological Mechanisms of the Chinese Patent Drug Kunling Pill for Improving Diminished Ovarian Reserve. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5929694. [PMID: 35668784 PMCID: PMC9167067 DOI: 10.1155/2022/5929694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 04/10/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022]
Abstract
Objective. To explore the pharmacological mechanism of a Chinese patent drug (Kunling Pill (KLP)) on improving diminished ovarian reserve based on proteomic analysis. Methods. A total of 18 patients divided into three groups (the normal ovary reserve (NOR), diminished ovary reserve (DOR), and KLP groups) undergoing assisted reproductive technology by standard ovarian stimulation protocols were recruited to collect follicular fluid. Data-independent acquisition mass spectrometry was used to identify differentially expressed proteins by nano-LC-MS/MS. Bioinformatic analysis was conducted to predict the functions and pathways of the identified proteins. Clinical, hormonal, and biochemical parameters were also analyzed in the three groups. Results. A total of 144 differentially expressed proteins were screened out, including 56 proteins that were downregulated and 88 proteins that were upregulated in the DOR group compared with the NOR group, while 27 proteins were shared in the KLP-treated group. Among them, 10 proteins were upregulated and 17 proteins were downregulated in the KLP-treated group compared with the DOR group. The most enriched biological processes accounted for 28 GO terms, including cellular process, biological regulation, metabolic process, and regulation of biological process. Significant pathways were associated with fatty acid elongation, fatty acid degradation, fatty acid metabolism, nicotinate and nicotinamide metabolism, and valine, leucine, and isoleucine degradation. Conclusion. Our study provides the proteome profiles of human follicular fluid from DOR patients treated by KLP. Functional analyses of proteome datasets revealed that core proteins (SAA1, MIF, and PRDX5) and related pathways (fatty acid metabolism, nicotinate and nicotinamide metabolism, and tyrosine and purine metabolism) are possible pharmacological mechanisms through which KLP improves DOR. Therefore, these findings may help better understand the complex mechanisms through which DOR is treated by the Chinese patent drug KLP.
Collapse
|
15
|
Yu Z, Gao J, Zhang X, Peng Y, Wei W, Xu J, Li Z, Wang C, Zhou M, Tian X, Feng L, Huo X, Liu M, Ye M, Guo DA, Ma X. Characterization of a small-molecule inhibitor targeting NEMO/IKKβ to suppress colorectal cancer growth. Signal Transduct Target Ther 2022; 7:71. [PMID: 35260565 PMCID: PMC8904520 DOI: 10.1038/s41392-022-00888-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
Abstract
NEMO/IKKβ complex is a central regulator of NF-κB signaling pathway, its dissociation has been considered to be an attractive therapeutic target. Herein, using a combined strategy of molecular pharmacological phenotyping, proteomics and bioinformatics analysis, Shikonin (SHK) is identified as a potential inhibitor of the IKKβ/NEMO complex. It destabilizes IKKβ/NEMO complex with IC50 of 174 nM, thereby significantly impairing the proliferation of colorectal cancer cells by suppressing the NF-κB pathway in vitro and in vivo. In addition, we also elucidated the potential target sites of SHK in the NEMO/IKKβ complex. Our study provides some new insights for the development of potent small-molecule PPI inhibitors.
Collapse
Affiliation(s)
- Zhenlong Yu
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, 116000, China
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Jian Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, China
| | - Xiaolei Zhang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Yulin Peng
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, 116000, China
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Wenlong Wei
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jianrong Xu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Zhenwei Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chao Wang
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Meirong Zhou
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Xiangge Tian
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, 116000, China
| | - Lei Feng
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, 116000, China
| | - Xiaokui Huo
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, 116000, China
| | - Min Liu
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
- Neurology Department, Dalian University Affiliated Xinhua Hospital, Dalian, 116021, China
| | - Mingliang Ye
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
| | - De-An Guo
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Xiaochi Ma
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, 116000, China.
- College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
16
|
Peroxiredoxin-5 Knockdown Accelerates Pressure Overload-Induced Cardiac Hypertrophy in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5067544. [PMID: 35132351 PMCID: PMC8817848 DOI: 10.1155/2022/5067544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 08/19/2021] [Accepted: 12/21/2021] [Indexed: 11/29/2022]
Abstract
A recent study showed that peroxiredoxins (Prxs) play an important role in the development of pathological cardiac hypertrophy. However, the involvement of Prx5 in cardiac hypertrophy remains unclear. Therefore, this study is aimed at investigating the role and mechanisms of Prx5 in pathological cardiac hypertrophy and dysfunction. Transverse aortic constriction (TAC) surgery was performed to establish a pressure overload-induced cardiac hypertrophy model. In this study, we found that Prx5 expression was upregulated in hypertrophic hearts and cardiomyocytes. In addition, Prx5 knockdown accelerated pressure overload-induced cardiac hypertrophy and dysfunction in mice by activating oxidative stress and cardiomyocyte apoptosis. Importantly, heart deterioration caused by Prx5 knockdown was related to mitogen-activated protein kinase (MAPK) pathway activation. These findings suggest that Prx5 could be a novel target for treating cardiac hypertrophy and heart failure.
Collapse
|
17
|
Soliman AM, Mekkawy MH, Karam HM, Higgins M, Dinkova-Kostova AT, Ghorab MM. Novel iodinated quinazolinones bearing sulfonamide as new scaffold targeting radiation induced oxidative stress. Bioorg Med Chem Lett 2021; 42:128002. [PMID: 33811990 DOI: 10.1016/j.bmcl.2021.128002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/12/2020] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS) play an integral role in the pathogenesis of most diseases. This work presents the design and synthesis of fourteen new diiodoquinazolinone derivatives bearing benzenesulfonamide moiety with variable acetamide tail and evaluation of their ability to activate nuclear factor erythroid 2-related factor 2 (Nrf2) using its classical target NAD(P)H: quinone oxidoreductase 1 (NQO1) in Hepa1c1c7 murine hepatoma cells. The N-(2-chloropyridin-3-yl)-2-((6,8-diiodo-4-oxo-3-(4-sulfamoylphenyl)-3,4-dihydroquinazolin-2-yl)thio) acetamide 17 was the most potent NQO1 inducer (CD = 25 µM) with free radical scavenging activity (IC50 = 28 µM) and in vivo median lethal dose (LD50) of 500 mg/Kg. The possible radioprotective activity of compound 17 was evaluated in (7 Gy) irradiated mice. Compound 17 showed a reduction in radiation induced oxidative stress as evidenced by the lower levels of ROS, malondialdehyde (MDA) and NQO1 in liver tissues. Moreover, compound 17 showed improvement in the complete blood count (CBC) of irradiated mice and decreased mortality over 30 days following irradiation. Additionally, docking studies inside the Nrf2-binding site of Kelch-like ECH associated protein 1 (Keap1), the main negative regulator of Nrf2, confirmed that 17 revealed the same interactions with the key amino acids as those of the co-crystallized ligand. This study identifies 17 as a novel antioxidant that protects against the harmful effect of radiation.
Collapse
Affiliation(s)
- Aiten M Soliman
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Nasr City P.O. Box 29, Cairo 11765, Egypt
| | - Mai H Mekkawy
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Nasr City P.O. Box 29, Cairo 11765, Egypt
| | - Heba M Karam
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Nasr City P.O. Box 29, Cairo 11765, Egypt
| | - Maureen Higgins
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, UK; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mostafa M Ghorab
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Nasr City P.O. Box 29, Cairo 11765, Egypt.
| |
Collapse
|
18
|
Bolduc J, Koruza K, Luo T, Malo Pueyo J, Vo TN, Ezeriņa D, Messens J. Peroxiredoxins wear many hats: Factors that fashion their peroxide sensing personalities. Redox Biol 2021; 42:101959. [PMID: 33895094 PMCID: PMC8113037 DOI: 10.1016/j.redox.2021.101959] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/29/2020] [Revised: 03/07/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Peroxiredoxins (Prdxs) sense and assess peroxide levels, and signal through protein interactions. Understanding the role of the multiple structural and post-translational modification (PTM) layers that tunes the peroxiredoxin specificities is still a challenge. In this review, we give a tabulated overview on what is known about human and bacterial peroxiredoxins with a focus on structure, PTMs, and protein-protein interactions. Armed with numerous cellular and atomic level experimental techniques, we look at the future and ask ourselves what is still needed to give us a clearer view on the cellular operating power of Prdxs in both stress and non-stress conditions.
Collapse
Affiliation(s)
- Jesalyn Bolduc
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Katarina Koruza
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Ting Luo
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Julia Malo Pueyo
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Trung Nghia Vo
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Daria Ezeriņa
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Joris Messens
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium.
| |
Collapse
|
19
|
Wang Q, Xu L, Wang G, Chen L, Li C, Jiang X, Gao H, Yang B, Tian W. Prognostic and clinicopathological significance of NRF2 expression in non-small cell lung cancer: A meta-analysis. PLoS One 2020; 15:e0241241. [PMID: 33186371 PMCID: PMC7665804 DOI: 10.1371/journal.pone.0241241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/09/2020] [Accepted: 10/09/2020] [Indexed: 01/14/2023] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) functions as a transcription factor and regulates a wide array of antioxidant and stress-responsive genes. NRF2 has been widely implicated in different types of cancers, but only limited studies concerning the relationship between NRF2 expression and tumour invasion or prognosis in lung cancer. Therefore, we conducted a meta-analysis to determine the prognostic value of NRF2 in patients with non-small cell lung cancer (NSCLC). The relationship between NRF2 expression in NSCLC patients and clinicopathological features was also investigated. Overall survival (OS) and treatment response rate were evaluated using STATA software. Twenty eligible articles with 2530 lung cancer patients were included in this meta-analysis. The results revealed that high expression level of NRF2 was associated with pathologic distant metastasis (odds ratio (OR) = 2.64, 95% confidence interval (CI) 1.62-4.31; P < 0.001), lymph node metastasis (OR = 2.14, 95% CI: 1.53-3.00; P < 0.001), and tumour node metastasis (TNM) stage (OR = 1.95, 95% CI: 1.52-2.49, P < 0.001). High NRF2 expression was associated with low treatment response rate in platinum-based chemotherapy (HR = 0.11, 95% CI 0.02-0.51; P = 0.005). High expression level of NRF2 is predictive for poor overall survival rate (HR = 1.86, 95% CI 1.44-2.41, P < 0.001) and poor progression-free survival (PFS) (HR = 2.27, 95% CI 1.26-4.09, P = 0.006). Compared to patients with a low level of NRF2 expression, patients with high NRF2 expression levels were associated with worse OS and PFS when given the chemotherapy or EGFR-TKI. Together, our meta-analysis results suggest that NRF2 can act as a potential indicator of NSCLC tumour aggressiveness and help the prognosis and design of a better treatment strategy for NSCLC patients.
Collapse
Affiliation(s)
- Qingsong Wang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Liang Xu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Gang Wang
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lei Chen
- Department of Otolaryngology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Changping Li
- Department of Health Statistics, College of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiangli Jiang
- Department of Thoracic Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hai Gao
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Bing Yang
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Weiping Tian
- Tianjin Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|