1
|
Zhang X, Yang L, Feng K, Zhang H, Chen Y, Li W, Wang X, Zhang M, Wu Y, Wei S, Zheng Y, Meng G, Meng W, Chen X, Tang J. Shuxuening injection improves myocardial injury after myocardial infarction by regulating macrophage polarization via the TLR4/NF-κB and PI3K/Akt signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156418. [PMID: 39879705 DOI: 10.1016/j.phymed.2025.156418] [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: 11/16/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Macrophage activation and polarization play pivotal roles in the inflammatory response and myocardial injury associated with myocardial infarction (MI). Modulating macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype is a promising therapeutic approach for MI. Shuxuening injection (SXNI) is extensively utilized in clinical settings for MI treatment and has demonstrated therapeutic efficacy. However, the effects of SXNI on macrophage polarization post-MI and its underlying mechanisms remain insufficiently understood. AIM OF THE STUDY This study is aimed to evaluate the effects of SXNI on macrophage polarization following MI and to elucidate its potential mechanisms of action. METHODS A rat model of MI was established by ligation of the left anterior descending coronary artery. The cardioprotective effects of SXNI were assessed through echocardiography, TTC staining, Masson's trichrome staining, HE staining, TUNEL staining, and western blotting (WB). Macrophage polarization was evaluated using ELISA, immunofluorescence staining, and WB. An in vitro model of oxygen-glucose deprivation (OGD) was utilized to simulate MI in macrophages, and qRT-PCR was employed to examine M1/M2 polarization markers. UPLC-Q-TOF/MS was used to identify active components in SXNI. Network pharmacology analysis and molecular docking were utilized to predict the key targets and pathways, which were subsequently validated through WB and immunohistochemistry. RESULTS SXNI improved cardiac function, reduced infarct size, and attenuated myocardial tissue damage and apoptosis in MI rats. Staining analyses indicated a reduction in M1 macrophages (CD86+/CD68+) and an increase in M2 macrophages (CD206+/CD68+) in SXNI-treated animals. In vivo and in vitro experiments demonstrated that SXNI decreased M1 markers and pro-inflammatory cytokines levels while increasing M2 markers and the production of anti-inflammatory and pro-angiogenic cytokines. UPLC-Q-TOF/MS analysis identified 18 active components in SXNI. Network pharmacology analysis and molecular docking implicated the TLR4/NF-κB and PI3K/Akt pathways as central mechanisms, which were further confirmed by WB and immunohistochemistry. SXNI inhibited the expression of TLR4 and phosphorylated NF-κB while enhancing phosphorylated PI3 K and Akt levels. CONCLUSIONS SXNI modulates the TLR4/NF-κB and PI3K/Akt signaling pathways to promote the polarization of macrophages from the M1 to the M2 phenotype, thereby alleviating myocardial inflammation and injury. These findings provide a scientific basis for the clinical application of SXNI in MI management, and establish a scientific foundation for exploring novel therapeutic strategies for cardiovascular diseases based on macrophage polarization.
Collapse
Affiliation(s)
- Xiaoshuai Zhang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Liuqing Yang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China.
| | - Kairui Feng
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Hui Zhang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China
| | - Yulong Chen
- College of pharmaceutical engineering of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Weixia Li
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Xiaoyan Wang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Mingliang Zhang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China
| | - Yali Wu
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China
| | - Shiting Wei
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Yajuan Zheng
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Gaoquan Meng
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Weiting Meng
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Xiaofei Chen
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China.
| | - Jinfa Tang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China.
| |
Collapse
|
2
|
Erber L, Groehler AS, Cyuzuzo CI, Baker-Wainwright J, Maskey RS, Li L, Machida YJ, Tretyakova N. SPRTN metalloprotease participates in repair of ROS-mediated DNA-protein crosslinks. Sci Rep 2024; 14:30919. [PMID: 39730693 DOI: 10.1038/s41598-024-81799-9] [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: 09/19/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
Exposure to reactive oxygen species (ROS) can induce DNA-protein crosslinks (DPCs), unusually bulky DNA lesions that block replication and transcription and play a role in aging, cancer, cardiovascular disease, and neurodegenerative disorders. Repair of DPCs depends on the coordinated efforts of proteases and DNA repair enzymes to cleave the protein component of the lesion to smaller DNA-peptide crosslinks which can be processed by tyrosyl-DNA phosphodiesterases 1 and 2, nucleotide excision and homologous recombination repair pathways. DNA-dependent metalloprotease SPRTN plays a role in DPC repair, and SPRTN-deficient mice exhibit an accelerated aging phenotype and develop liver cancer early in life. We investigated the role of the SPRTN enzyme in the repair of DPCs produced by a free radical mechanism. Sprtn-deficient MEF cells treated with ionizing radiation had higher levels of total DPCs and exhibited greater sensitivity upon exposure to hydrogen peroxide and other crosslinking agents including cisplatin, phosphoramide mustard, and 1,2,3,4-diepoxybutane. Using a sensitive and accurate nanoLC-ESI+-MS/MS assay, we specifically measured the radical-induced crosslinking of thymidine in DNA crosslinking of thymidine in DNA to tyrosine in proteins (dT-Tyr) in the tissues of SPRTN hypomorphic (SprtnH/H) and wild type mice. Genomic DNA isolated from the tissues of SPRTN hypomorphic (SprtnH/H) mice exhibited higher levels of dT-Tyr in the liver, brain, heart, and kidney than wild-type animals. Overall, our results are consistent with the understanding that SPRTN has a role in maintaining genomic integrity upon exposure to ionizing radiation and endogenous reactive oxygen species.
Collapse
Affiliation(s)
- Luke Erber
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, 66045, USA
| | - Arnold S Groehler
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Cesar I Cyuzuzo
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | | | - Reeja S Maskey
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lei Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yuichi J Machida
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Natalia Tretyakova
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
3
|
Soldatov V, Venediktov A, Belykh A, Piavchenko G, Naimzada MD, Ogneva N, Kartashkina N, Bushueva O. Chaperones vs. oxidative stress in the pathobiology of ischemic stroke. Front Mol Neurosci 2024; 17:1513084. [PMID: 39723236 PMCID: PMC11668803 DOI: 10.3389/fnmol.2024.1513084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
As many proteins prioritize functionality over constancy of structure, a proteome is the shortest stave in the Liebig's barrel of cell sustainability. In this regard, both prokaryotes and eukaryotes possess abundant machinery supporting the quality of the proteome in healthy and stressful conditions. This machinery, namely chaperones, assists in folding, refolding, and the utilization of client proteins. The functions of chaperones are especially important for brain cells, which are highly sophisticated in terms of structural and functional organization. Molecular chaperones are known to exert beneficial effects in many brain diseases including one of the most threatening and widespread brain pathologies, ischemic stroke. However, whether and how they exert the antioxidant defense in stroke remains unclear. Herein, we discuss the chaperones shown to fight oxidative stress and the mechanisms of their antioxidant action. In ischemic stroke, during intense production of free radicals, molecular chaperones preserve the proteome by interacting with oxidized proteins, regulating imbalanced mitochondrial function, and directly fighting oxidative stress. For instance, cells recruit Hsp60 and Hsp70 to provide proper folding of newly synthesized proteins-these factors are required for early ischemic response and to refold damaged polypeptides. Additionally, Hsp70 upregulates some dedicated antioxidant pathways such as FOXO3 signaling. Small HSPs decrease oxidative stress via attenuation of mitochondrial function through their involvement in the regulation of Nrf- (Hsp22), Akt and Hippo (Hsp27) signaling pathways as well as mitophagy (Hsp27, Hsp22). A similar function has also been proposed for the Sigma-1 receptor, contributing to the regulation of mitochondrial function. Some chaperones can prevent excessive formation of reactive oxygen species whereas Hsp90 is suggested to be responsible for pro-oxidant effects in ischemic stroke. Finally, heat-resistant obscure proteins (Hero) are able to shield client proteins, thus preventing their possible over oxidation.
Collapse
Affiliation(s)
- Vladislav Soldatov
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| | - Artem Venediktov
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Andrei Belykh
- Pathophysiology Department, Kursk State Medical University, Kursk, Russia
- Research Institute of General Pathology, Kursk State Medical University, Kursk, Russia
| | - Gennadii Piavchenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Mukhammad David Naimzada
- Research Institute of Experimental Medicine, Kursk State Medical University, Kursk, Russia
- Laboratory of Public Health Indicators Analysis and Health Digitalization, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Nastasya Ogneva
- Scientific Center of Biomedical Technologies, Federal Medical and Biological Agency of Russia, Moscow, Russia
| | - Natalia Kartashkina
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Olga Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russia
| |
Collapse
|
4
|
Li M, Wu J, Yang T, Zhao Y, Ren P, Chang L, Shi P, Yang J, Liu Y, Li X, Wang P, Cao Y. Engineered Biomimetic Nanoparticles-Mediated Targeting Delivery of Allicin Against Myocardial Ischemia-Reperfusion Injury by Inhibiting Ferroptosis. Int J Nanomedicine 2024; 19:11275-11292. [PMID: 39524923 PMCID: PMC11550785 DOI: 10.2147/ijn.s478276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Background Cardiac microvascular damage is substantially related with the onset of myocardial ischaemia-reperfusion (IR) injury. Reportedly, allicin (AL) effectively protects the cardiac microvascular system from IR injury. However, the unsatisfactory therapeutic efficacy of current drugs and insufficient drug delivery to the damaged heart are major concerns. Here, inspired by the natural interaction between neutrophils and inflamed cardiac microvascular endothelial cells (CMECs), a neutrophil membrane-camouflaged nanoparticle for non-invasive active-targeting therapy for IR injury by improving drug delivery to the injured heart is constructed. Methods In this study, we engineered mesoporous silica nanoparticles (MSNs) coated with a neutrophil membrane to act as a drug delivery system, encapsulating AL. The potential of the nanoparticles (named AL@MSNs@NM) for specific targeting of infarcted myocardium was assessed using small animal vivo imaging system. The cardiac function of AL@MSNs@NM after treatment was evaluated by Animal Ultrasound Imaging system, HE staining, and Laser Speckle Imaging System. The therapeutic mechanism was analyzed by ELISA kits, immunofluorescence, and PCR. Results We discovered that AL@MSNs@NM significantly improves cardiac function index, reduced infarct size and fibrosis, increased vascular perfusion in ischemic areas, and also promoted the function of CMECs, including migration, tube formation, shear stress adaptation, and nitric oxide production. Further research revealed that AL@MSNs@NM have cardio-protective functions in IR rats by inhibiting CMEC ferroptosis and increasing platelet endothelial cell adhesion molecule-1 (PECAM-1) expression. Conclusion Our results indicated that AL@MSNs@NM significantly reversed CMEC ferroptosis and increased PECAM-1 expression, enhanced cardiac function, and reduced myocardial infarction size. Therefore, this strategy demonstrates that engineered biomimetic nanotechnology effectively delivers AL for targeted therapy of myocardial infarction.
Collapse
Affiliation(s)
- Minghui Li
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Jiabi Wu
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Tao Yang
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Yuhang Zhao
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Ping Ren
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Lingling Chang
- Department of Physiology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Pilong Shi
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Jing Yang
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Yuhang Liu
- Department of Physiology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Xiaolei Li
- Department of Pathology, Jiangsu College of Nursing, Jiangsu, 223003, People’s Republic of China
| | - Peng Wang
- Department of Physiology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| | - Yonggang Cao
- Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China
| |
Collapse
|
5
|
Kong W, Zhao Y, Dai X, You C. Methodologies for the detection and sequencing of the epigenetic-like oxidative DNA modification, 8-oxo-7,8-dihydroguanine. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 794:108516. [PMID: 39486616 DOI: 10.1016/j.mrrev.2024.108516] [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: 05/16/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
The human genome is constantly threatened by endogenous and environmental DNA damaging agents that can induce a variety of chemically modified DNA lesions including 8-oxo-7,8-dihydroguanine (OG). Increasing evidence has indicated that OG is not only a biomarker for oxidative DNA damage but also a novel epigenetic-like modification involved in regulation of gene expression in mammalian cells. Here we summarize the recent progress in OG research focusing on the following points: (i) the mechanism of OG production in organisms and its biological consequences in cells, (ii) the accurate identification of OG in low-abundance genomes and complex biological backgrounds, (iii) the development of OG sequencing methods. These studies will be helpful for further understanding of the molecular mechanisms of OG-induced mutagenesis and its potential roles in human development and diseases such as cancer.
Collapse
Affiliation(s)
- Weiheng Kong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Molecular Science and Biomedicine Laboratory, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China; College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong 273165, China
| | - Yingqi Zhao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Molecular Science and Biomedicine Laboratory, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Xiaoxia Dai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Molecular Science and Biomedicine Laboratory, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.
| | - Changjun You
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Molecular Science and Biomedicine Laboratory, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.
| |
Collapse
|
6
|
Hurben AK, Zhang Q, Galligan JJ, Tretyakova N, Erber L. Endogenous Cellular Metabolite Methylglyoxal Induces DNA-Protein Cross-Links in Living Cells. ACS Chem Biol 2024; 19:1291-1302. [PMID: 38752800 PMCID: PMC11353540 DOI: 10.1021/acschembio.4c00100] [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] [Indexed: 06/22/2024]
Abstract
Methylglyoxal (MGO) is an electrophilic α-oxoaldehyde generated endogenously through metabolism of carbohydrates and exogenously due to autoxidation of sugars, degradation of lipids, and fermentation during food and drink processing. MGO can react with nucleophilic sites within proteins and DNA to form covalent adducts. MGO-induced advanced glycation end-products such as protein and DNA adducts are thought to be involved in oxidative stress, inflammation, diabetes, cancer, renal failure, and neurodegenerative diseases. Additionally, MGO has been hypothesized to form toxic DNA-protein cross-links (DPC), but the identities of proteins participating in such cross-linking in cells have not been determined. In the present work, we quantified DPC formation in human cells exposed to MGO and identified proteins trapped on DNA upon MGO exposure using mass spectrometry-based proteomics. A total of 265 proteins were found to participate in MGO-derived DPC formation including gene products engaged in telomere organization, nucleosome assembly, and gene expression. In vitro experiments confirmed DPC formation between DNA and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), as well as histone proteins H3.1 and H4. Collectively, our study provides the first evidence for MGO-mediated DNA-protein cross-linking in living cells, prompting future studies regarding the relevance of these toxic lesions in cancer, diabetes, and other diseases linked to elevated MGO levels.
Collapse
Affiliation(s)
- Alexander K. Hurben
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States; Present Address: Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Qi Zhang
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - James J. Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Natalia Tretyakova
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Luke Erber
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
7
|
Yang F, Shen C. Sodium Danshensu Cream Promotes the Healing of Pressure Ulcers in Mice through the Nrf2/HO-1 and NF-κB Pathways. Pharmaceuticals (Basel) 2022; 15:ph15121548. [PMID: 36558999 PMCID: PMC9783848 DOI: 10.3390/ph15121548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
On the basis of the mice pressure ulcers (PU) model, the protective effect and potential mechanism of sodium Danshensu (SDSS) cream against PU were investigated. The mice were randomly divided into three groups: the negative control group (cream without 0.5 g SDSS), the SDSS group (cream containing 0.5 g SDSS), and the positive group (0.5 g Hirudoid®). After 7 and 14 days of ointment application, the wound-healing rate of the SDSS and positive groups was significantly higher than that of the control group (p < 0.05). The results of hematoxylin−eosin staining also indicated that SDSS has the potential to promote the healing of PU. In addition, the serum IL-6, IL-1β, TNF-α, and MDA levels decreased significantly (p < 0.01) after 14 days of SDSS treatment, while the SOD, CAT, and GSH-Px activities increased significantly (p < 0.01). In addition, SDSS cream was able to significantly increase the expression of Nrf2, HO-1, GCLM, NQO1, NF-κB p65, NF-κB p50, IKKα, and IKKβ while decreasing the expression of Keap1 and IκBαin the Nrf2/HO-1 and NF-κB pathways. Our research will provide a foundation for the future clinical prevention and treatment of PU with SDSS cream.
Collapse
Affiliation(s)
- Fei Yang
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou 310008, China
| | - Cuizhen Shen
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Correspondence:
| |
Collapse
|
8
|
Changes in metabolic profiling of whiteleg shrimp (Penaeus vannamei) under hypoxic stress. J Invertebr Pathol 2022; 193:107798. [PMID: 35843291 DOI: 10.1016/j.jip.2022.107798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/13/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022]
Abstract
Hypoxia is a common concern in shrimp aquaculture, affecting growth and survival. Although recent studies have revealed important insights into hypoxia in shrimp and crustaceans, knowledge gaps remain regarding this stressor at the molecular level. In the present study, a gas chromatography-mass spectrometry (GC-MS)-based metabolomics approach was employed to characterize the metabolic signatures and pathways underlying responses of Pacific white shrimp (Penaeus vannamei) to hypoxia and to identify associated candidate biomarkers. We compared metabolite profiles of shrimp haemolymph before (0 h) and after exposure to hypoxia (1 & 2 h). Dissolved oxygen levels were maintained above 85 % saturation in the control and before hypoxia, and 15 % saturation in the hypoxic stress treatment. Results showed 44 metabolites in shrimp haemolymph that were significantly different between before and after hypoxia exposure. These metabolites were energy-related metabolites (e.g., intermediates of citric acid cycle, lactic acid, alanine), fatty acids and amino acids. Pathway analysis revealed 17 pathways that were significantly affected by hypoxia. The changes in metabolites and pathways indicate a shift from aerobic to anaerobic metabolism, disturbance in amino acid metabolism, osmoregulation, oxidative damage and Warburg effect-like response caused by hypoxic stress. Among the altered metabolites, lactic acid was most different between before and after hypoxia exposure and had the highest accurate value for biomarker identification. Future investigations may validate this molecule as a stress biomarker in aquaculture. This study contributes to a better understanding of hypoxia in shrimp and crustaceans at the metabolic level and provides a base for future metabolomics investigations on hypoxia.
Collapse
|
9
|
Ghodke PP, Matse JH, Dawson S, Guengerich FP. Nucleophilic Thiol Proteins Bind Covalently to Abasic Sites in DNA. Chem Res Toxicol 2022; 35:1805-1808. [PMID: 35482010 DOI: 10.1021/acs.chemrestox.2c00068] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In the course of studies on the enhancement of 1,2-dibromoethane-induced DNA base pair mutations by O6-alkylguanine-DNA alkyltransferase (AGT, MGMT), we discovered the facile reaction of AGT with an abasic site in DNA, leading to covalent cross-linking. The binding of AGT differs from the mechanism reported for the protein HMCES; instead it appears to involve formation of a stable thioglycoside. Facile cross-linking was also observed with the protease papain, which like AGT has a low pKa cysteine, and the tripeptide glutathione.
Collapse
Affiliation(s)
- Pratibha P Ghodke
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Johannes H Matse
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Scott Dawson
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| |
Collapse
|
10
|
Stoneley M, Harvey RF, Mulroney TE, Mordue R, Jukes-Jones R, Cain K, Lilley KS, Sawarkar R, Willis AE. Unresolved stalled ribosome complexes restrict cell-cycle progression after genotoxic stress. Mol Cell 2022; 82:1557-1572.e7. [PMID: 35180429 PMCID: PMC9098122 DOI: 10.1016/j.molcel.2022.01.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 01/01/2023]
Abstract
During the translation surveillance mechanism known as ribosome-associated quality control, the ASC-1 complex (ASCC) disassembles ribosomes stalled on the mRNA. Here, we show that there are two distinct classes of stalled ribosome. Ribosomes stalled by translation elongation inhibitors or methylated mRNA are short lived in human cells because they are split by the ASCC. In contrast, although ultraviolet light and 4-nitroquinoline 1-oxide induce ribosome stalling by damaging mRNA, and the ASCC is recruited to these stalled ribosomes, we found that they are refractory to the ASCC. Consequently, unresolved UV- and 4NQO-stalled ribosomes persist in human cells. We show that ribosome stalling activates cell-cycle arrest, partly through ZAK-p38MAPK signaling, and that this cell-cycle delay is prolonged when the ASCC cannot resolve stalled ribosomes. Thus, we propose that the sensitivity of stalled ribosomes to the ASCC influences the kinetics of stall resolution, which in turn controls the adaptive stress response.
Collapse
Affiliation(s)
- Mark Stoneley
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.
| | - Robert F Harvey
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Thomas E Mulroney
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Ryan Mordue
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Rebekah Jukes-Jones
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Kelvin Cain
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Kathryn S Lilley
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre, Department of Biochemistry, University of Cambridge, Cambridge CB2 1QR, UK
| | - Ritwick Sawarkar
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Anne E Willis
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.
| |
Collapse
|
11
|
Ghodke PP, Gonzalez-Vasquez G, Wang H, Johnson KM, Sedgeman CA, Guengerich FP. Enzymatic bypass of an N 6-deoxyadenosine DNA-ethylene dibromide-peptide cross-link by translesion DNA polymerases. J Biol Chem 2021; 296:100444. [PMID: 33617883 PMCID: PMC8024977 DOI: 10.1016/j.jbc.2021.100444] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/25/2022] Open
Abstract
Unrepaired DNA-protein cross-links, due to their bulky nature, can stall replication forks and result in genome instability. Large DNA-protein cross-links can be cleaved into DNA-peptide cross-links, but the extent to which these smaller fragments disrupt normal replication is not clear. Ethylene dibromide (1,2-dibromoethane) is a known carcinogen that can cross-link the repair protein O6-alkylguanine-DNA alkyltransferase (AGT) to the N6 position of deoxyadenosine (dA) in DNA, as well as four other positions in DNA. We investigated the effect of a 15-mer peptide from the active site of AGT, cross-linked to the N6 position of dA, on DNA replication by human translesion synthesis DNA polymerases (Pols) η, ⍳, and κ. The peptide-DNA cross-link was bypassed by the three polymerases at different rates. In steady-state kinetics, the specificity constant (kcat/Km) for incorporation of the correct nucleotide opposite to the adduct decreased by 220-fold with Pol κ, tenfold with pol η, and not at all with Pol ⍳. Pol η incorporated all four nucleotides across from the lesion, with the preference dT > dC > dA > dG, while Pol ⍳ and κ only incorporated the correct nucleotide. However, LC-MS/MS analysis of the primer-template extension product revealed error-free bypass of the cross-linked 15-mer peptide by Pol η. We conclude that a bulky 15-mer peptide cross-linked to the N6 position of dA can retard polymerization and cause miscoding but that overall fidelity is not compromised because only correct pairs are extended.
Collapse
Affiliation(s)
- Pratibha P Ghodke
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | - Hui Wang
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kevin M Johnson
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Carl A Sedgeman
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| |
Collapse
|
12
|
He F, Zheng G, Hou J, Hu Q, Ling Q, Wu G, Zhao H, Yang J, Wang Y, Jiang L, Tang W, Yang Z. N-acetylcysteine alleviates post-resuscitation myocardial dysfunction and improves survival outcomes via partly inhibiting NLRP3 inflammasome induced-pyroptosis. JOURNAL OF INFLAMMATION-LONDON 2020; 17:25. [PMID: 32782443 PMCID: PMC7409674 DOI: 10.1186/s12950-020-00255-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
Background NOD-like receptor 3 (NLRP3) inflammasome is necessary to initiate acute sterile inflammation. Increasing evidence indicates the activation of NLRP3 inflammasome induced pyroptosis is closely related to reactive oxygen species (ROS) in the sterile inflammatory response triggered by ischemia/reperfusion (I/R) injury. N-acetylcysteine (NAC) is an antioxidant and plays a protective role in local myocardial I/R injury, while its effect on post-resuscitation myocardial dysfunction, as well as its mechanisms, remain elusive. In this study, we aimed to investigate the effect of NAC on post-resuscitation myocardial dysfunction in a cardiac arrest rat model, and whether its underlying mechanism may be linked to ROS and NLRP3 inflammasome-induced pyroptosis. Methods The rats were randomized into three groups: (1) sham group, (2) cardiopulmonary resuscitation (CPR) group, and (3) CPR + NAC group. CPR group and CPR + NAC group went through the induction of ventricular fibrillation (VF) and resuscitation. After return of spontaneous circulation (ROSC), rats in the CPR and CPR + NAC groups were again randomly divided into two subgroups, ROSC 6 h and ROSC 72 h, for further analysis. Hemodynamic measurements and myocardial function were measured by echocardiography, and western blot was used to detect the expression of proteins. Results Results showed that after treatment with NAC, there was significantly better myocardial function and survival duration; protein expression levels of NLRP3, adaptor apoptosis-associated speck-like protein (ASC), Cleaved-Caspase-1 and gasdermin D (GSDMD) in myocardial tissues were significantly decreased; and inflammatory cytokines levels were reduced. The marker of oxidative stress malondialdehyde (MDA) decreased and superoxide dismutase (SOD) increased with NAC treatment. Conclusions NAC improved myocardial dysfunction and prolonged animal survival duration in a rat model of cardiac arrest. Moreover, possibly by partly inhibiting ROS-mediated NLRP3 inflammasome-induced pryoptosis.
Collapse
Affiliation(s)
- Fenglian He
- The Second Hospital of Anhui Medical University, Hefei, 230032 China
| | - Guanghui Zheng
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120 China
| | - Jingying Hou
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120 China
| | - Qiaohua Hu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120 China
| | - Qin Ling
- Weil Institute of Emergency and Critical Care Research, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284 USA
| | - Gongfa Wu
- Weil Institute of Emergency and Critical Care Research, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284 USA
| | - Hui Zhao
- The Second Hospital of Anhui Medical University, Hefei, 230032 China
| | - Jin Yang
- The Second Hospital of Anhui Medical University, Hefei, 230032 China
| | - Yue Wang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120 China
| | - Longyuan Jiang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120 China
| | - Wanchun Tang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120 China.,Weil Institute of Emergency and Critical Care Research, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284 USA
| | - Zhengfei Yang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120 China.,Zeng Cheng District People's Hospital of Guang Zhou, Guangzhou, 511300 China
| |
Collapse
|
13
|
Effects of hypoxia and reoxygenation on intermediary metabolite homeostasis of marine bivalves Mytilus edulis and Crassostrea gigas. Comp Biochem Physiol A Mol Integr Physiol 2020; 242:110657. [DOI: 10.1016/j.cbpa.2020.110657] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 02/06/2023]
|
14
|
Xin G, Xu-Yong L, Shan H, Gang W, Zhen C, Ji-Jun L, Ping Y, Man-Hua C. SH2B1 protects cardiomyocytes from ischemia/reperfusion injury via the activation of the PI3K/AKT pathway. Int Immunopharmacol 2020; 83:105910. [PMID: 32222636 DOI: 10.1016/j.intimp.2019.105910] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/27/2019] [Accepted: 09/08/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Apoptosis, reactive oxidative stress (ROS) and inflammation act as the pivotal pathogenesis of myocardial ischemia/reperfusion (I/R) injury (MIRI). Our prior study and other investigation have demonstrated the participations of src homology 2 (SH2) B adaptor protein 1 (SH2B1) in ischemic injury and cardiac hypertrophy; whereas, the involvements of SH2B1 in MIRI and underlying mechanisms are completely unknown. METHOD In present study, MIRI model in vivo was induced by 30 min of ligation of LAD coronary artery and 24 h of reperfusion, and primary cultured cardiomyocytes were challenged with 2 h of hypoxia followed by 4 h of reoxygenation (H/R) to mimic MIRI in vitro. Adenovirus encoding for SH2B1 or GFP were pre-transfected into myocardium prior to MIRI both in vivo and in vitro. The myocardial damage, cardiac function, apoptosis, ROS and inflammation were evaluated systematically. Immunofluorescence staining and western blotting were alternatively performed to detect protein expression. RESULTS The results exhibited that H/R or I/R significantly reduced SH2B1 in cardiomyocytes, followed by impaired cell survival and function, which were strongly reversed after the adenovirus-mediated SH2B1 up-regulation. Meanwhile, I/R- and H/R-elevated inflammation, apoptosis and ROS were also alleviated by SH2B1 up-regulation. A mechanistic study suggested that the protective contributions of SH2B1 on H/R-suffered cardiomyocytes were based on the activation of the PI3K/AKT pathway. The abolishment of the PI3K/AKT via a pharmacological inhibitor (LY294002) repressed anti-H/R capabilities of SH2B1. CONCLUSION Therefore, SH2B1 prevents cardiomyocytes from inflammation, apoptosis and ROS in MIRI partially through the PI3K/AKT-dependent avenues. It may provide a novel therapeutic target for the treatment of MIRI.
Collapse
Affiliation(s)
- Guo Xin
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China
| | - Li Xu-Yong
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China
| | - Hu Shan
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China
| | - Wu Gang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Zhen
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China
| | - Liu Ji-Jun
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China
| | - Ye Ping
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China.
| | - Chen Man-Hua
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China.
| |
Collapse
|
15
|
Falfushynska H, Piontkivska H, Sokolova IM. Effects of intermittent hypoxia on cell survival and inflammatory responses in the intertidal marine bivalves Mytilus edulis and Crassostrea gigas. J Exp Biol 2020; 223:jeb217026. [PMID: 31953358 DOI: 10.1242/jeb.217026] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/10/2020] [Indexed: 12/12/2022]
Abstract
Hypoxia is a major stressor in estuarine and coastal habitats, leading to adverse effects in aquatic organisms. Estuarine bivalves such as blue mussels (Mytilus edulis) and Pacific oysters (Crassostrea gigas) can survive periodic oxygen deficiency but the molecular mechanisms that underlie cellular injury during hypoxia-reoxygenation are not well understood. We examined the molecular markers of autophagy, apoptosis and inflammation during short-term (1 day) and long-term (6 days) hypoxia and post-hypoxic recovery (1 h) in mussels and oysters by measuring the lysosomal membrane stability, activity of a key autophagic enzyme (cathepsin D) and mRNA expression of the genes involved in the cellular survival and inflammation, including caspase 2, 3 and 8, Bcl-2, BAX, TGF-β-activated kinase 1 (TAK1), nuclear factor kappa B1 (NF-κB) and NF-κB activating kinases IKKα and TBK1. Crassostrea gigas exhibited higher hypoxia tolerance, as well as blunted or delayed inflammatory and apoptotic response to hypoxia and reoxygenation as shown by the later onset and/or the lack of transcriptional activation of caspases, BAX and the inflammatory effector NF-κB, compared with M. edulis Long-term hypoxia resulted in upregulation of Bcl-2 in the oysters and mussels, implying activation of anti-apoptotic mechanisms. Our findings indicate the potential importance of the cell survival pathways in hypoxia tolerance of marine bivalves, and demonstrate the utility of the molecular markers of apoptosis and autophagy for the assessment of sublethal hypoxic stress in bivalve populations.
Collapse
Affiliation(s)
- Halina Falfushynska
- Department of Marine Biology, Institute of Biological Sciences, University of Rostock, 18055 Rostock, Germany
- Department of Human Health, Physical Rehabilitation and Vital Activity, Ternopil V. Hnatiuk National Pedagogical University, 46002 Ternopil, Ukraine
| | - Helen Piontkivska
- Department of Biological Sciences, Kent State University, Kent, OH 44243, USA
| | - Inna M Sokolova
- Department of Marine Biology, Institute of Biological Sciences, University of Rostock, 18055 Rostock, Germany
- Department of Maritime Systems, Interdisciplinary Faculty, University of Rostock, 18055 Rostock, Germany
| |
Collapse
|
16
|
Cong Y, Chi Q, Teng X, Li S. The Protection of Selenium Against Cadmium-Induced Mitochondrial Damage via the Cytochrome P450 in the Livers of Chicken. Biol Trace Elem Res 2019; 190:484-492. [PMID: 30392018 DOI: 10.1007/s12011-018-1557-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/23/2018] [Indexed: 12/14/2022]
Abstract
Cadmium (Cd) is a heavy metal in natural environment and has extreme toxicity. Selenium (Se) has protective effect against heavy metal-induced injury or oxidative stress. Cytochrome P450 (CYP450) enzymes are a family of hemoproteins primarily responsible for detoxification functions. In order to investigate whether CYP450 is related to the damage of livers caused by Cd exposure, we chose forty-eight 28-day-old healthy Hailan cocks for four groups: control group, Se group, Cd group, and Se + Cd group. After 90-day treatment, euthanized for experiment. Based on an established subchronic Cd poisoning model in chicken, this experiment was designed to detect mitochondrial structure, malondialdehyde (MDA), glutathione (GSH), DNA and protein crosslink (DPC) and protein carbonyl (PCO) content, the CYP450 and b5 contents, the aminopyrine-N-demethylase (AND), erythromycin N-demethylase (ERND), aniline 4-hydroxylase (AH) and NADPH-cytochrome C reducatase (CR) activities, and mRNA expression level in the livers. The present results indicated that the MDA content, PCO content, and DPC index in Cd group were higher than those observed in other three groups. Most of the mitochondrial structure is incomplete in Cd group. The contents of CYP450 and b5 were decreased in Cd group. The activities of AND, ERND, AH, and CR got reduced after Cd exposure, as observed in CYP450 gene expression. Our results showed that CYP450 system was involved in the entire process of injury and protection. This research provides a comprehensive evaluation of the oxidative stress effects of Cd related to CYP450 in chicken.
Collapse
Affiliation(s)
- Yimei Cong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Qianru Chi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
17
|
Novel Molecular Targets Participating in Myocardial Ischemia-Reperfusion Injury and Cardioprotection. Cardiol Res Pract 2019; 2019:6935147. [PMID: 31275641 PMCID: PMC6558612 DOI: 10.1155/2019/6935147] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022] Open
Abstract
Worldwide morbidity and mortality from acute myocardial infarction (AMI) and related heart failure remain high. While effective early reperfusion of the criminal coronary artery after a confirmed AMI is the typical treatment at present, collateral myocardial ischemia-reperfusion injury (MIRI) and pertinent cardioprotection are still challenging to address and have inadequately understood mechanisms. Therefore, unveiling the related novel molecular targets and networks participating in triggering and resisting the pathobiology of MIRI is a promising and valuable frontier. The present study specifically focuses on the recent MIRI advances that are supported by sophisticated bio-methodology in order to bring the poorly understood interrelationship among pro- and anti-MIRI participant molecules up to date, as well as to identify findings that may facilitate the further investigation of novel targets.
Collapse
|
18
|
Intrinsic Effects of Gold Nanoparticles on Oxygen-Glucose Deprivation/Reperfusion Injury in Rat Cortical Neurons. Neurochem Res 2019; 44:1549-1566. [PMID: 31093902 DOI: 10.1007/s11064-019-02776-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 01/14/2023]
Abstract
This study aimed to investigate the potential effects of gold nanoparticles (Au-NPs) on rat cortical neurons exposed to oxygen-glucose deprivation/reperfusion (OGD/R) and to elucidate the corresponding mechanisms. Primary rat cortical neurons were exposed to OGD/R, which is commonly used in vitro to mimic ischemic injury, and then treated with 5- or 20-nm Au-NPs. We then evaluated cell viability, apoptosis, oxidative stress, and mitochondrial respiration in these neurons. We found that 20-nm Au-NPs increased cell viability, alleviated neuronal apoptosis and oxidative stress, and improved mitochondrial respiration after OGD/R injury, while opposite effects were observed for 5-nm Au-NPs. In terms of the underlying mechanisms, we found that Au-NPs could regulate Akt signaling. Taken together, these results show that 20-nm Au-NPs can protect primary cortical neurons against OGD/R injury, possibly by decreasing apoptosis and oxidative stress, while activating Akt signaling and mitochondrial pathways. Our results suggest that Au-NPs may be potential therapeutic agents for ischemic stroke.
Collapse
|
19
|
Effects of hypoxia-reoxygenation stress on mitochondrial proteome and bioenergetics of the hypoxia-tolerant marine bivalve Crassostrea gigas. J Proteomics 2019; 194:99-111. [DOI: 10.1016/j.jprot.2018.12.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/03/2018] [Accepted: 12/10/2018] [Indexed: 12/21/2022]
|
20
|
Gong X, Liu H, Wang S, Liang S, Wang G. Exosomes derived from SDF1‐overexpressing mesenchymal stem cells inhibit ischemic myocardial cell apoptosis and promote cardiac endothelial microvascular regeneration in mice with myocardial infarction. J Cell Physiol 2019; 234:13878-13893. [PMID: 30720220 DOI: 10.1002/jcp.28070] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/14/2018] [Accepted: 12/18/2018] [Indexed: 01/23/2023]
Affiliation(s)
- Xu‐He Gong
- Department of Cardiology Beijing Friendship Hospital, Capital Medical University Beijing China
| | - Hui Liu
- Department of Cardiology State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Si‐Jia Wang
- Department of Emergency Beijing Friendship Hospital, Capital Medical University Beijing China
| | - Si‐Wen Liang
- Department of Cardiology Beijing Friendship Hospital, Capital Medical University Beijing China
| | - Guo‐Gan Wang
- Department of Cardiology State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| |
Collapse
|
21
|
Effects of Ambient Atmospheric PM2.5, 1-Nitropyrene and 9-Nitroanthracene on DNA Damage and Oxidative Stress in Hearts of Rats. Cardiovasc Toxicol 2018; 19:178-190. [DOI: 10.1007/s12012-018-9488-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
22
|
Marine natural products for multi-targeted cancer treatment: A future insight. Biomed Pharmacother 2018; 105:233-245. [PMID: 29859466 DOI: 10.1016/j.biopha.2018.05.142] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/26/2018] [Accepted: 05/28/2018] [Indexed: 12/28/2022] Open
Abstract
Cancer is world's second largest alarming disease, which involves abnormal cell growth and have potential to spread to other parts of the body. Most of the available anticancer drugs are designed to act on specific targets by altering the activity of involved transporters and genes. As cancer cells exhibit complex cellular machinery, the regeneration of cancer tissues and chemo resistance towards the therapy has been the main obstacle in cancer treatment. This fact encourages the researchers to explore the multitargeted use of existing medicines to overcome the shortcomings of chemotherapy for alternative and safer treatment strategies. Recent developments in genomics-proteomics and an understanding of the molecular pharmacology of cancer have also challenged researchers to come up with target-based drugs. The literature supports the evidence of natural compounds exhibiting antioxidant, antimitotic, anti-inflammatory, antibiotic as well as anticancer activity. In this review, we have selected marine sponges as a prolific source of bioactive compounds which can be explored for their possible use in cancer and have tried to link their role in cancer pathway. To prove this, we revisited the literature for the selection of cancer genes for the multitargeted use of existing drugs and natural products. We used Cytoscape network analysis and Search tool for retrieval of interacting genes/ proteins (STRING) to study the possible interactions to show the links between the antioxidants, antibiotics, anti-inflammatory and antimitotic agents and their targets for their possible use in cancer. We included total 78 pathways, their genes and natural compounds from the above four pharmacological classes used in cancer treatment for multitargeted approach. Based on the Cytoscape network analysis results, we shortlist 22 genes based on their average shortest path length connecting one node to all other nodes in a network. These selected genes are CDKN2A, FH, VHL, STK11, SUFU, RB1, MEN1, HRPT2, EXT1, 2, CDK4, p14, p16, TSC1, 2, AXIN2, SDBH C, D, NF1, 2, BHD, PTCH, GPC3, CYLD and WT1. The selected genes were analysed using STRING for their protein-protein interactions. Based on the above findings, we propose the selected genes to be considered as major targets and are suggested to be studied for discovering marine natural products as drug lead in cancer treatment.
Collapse
|